WO2020074001A1 - Methods and Compositions for Editing RNAs - Google Patents

Methods and Compositions for Editing RNAs Download PDF

Info

Publication number
WO2020074001A1
WO2020074001A1 PCT/CN2019/110782 CN2019110782W WO2020074001A1 WO 2020074001 A1 WO2020074001 A1 WO 2020074001A1 CN 2019110782 W CN2019110782 W CN 2019110782W WO 2020074001 A1 WO2020074001 A1 WO 2020074001A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
target
drna
editing
target rna
Prior art date
Application number
PCT/CN2019/110782
Other languages
English (en)
French (fr)
Inventor
Wensheng Wei
Liang QU
Zongyi YI
Shiyou ZHU
Chunhui WANG
Zhongzheng CAO
Zhuo ZHOU
Pengfei YUAN
Original Assignee
Peking University
Edigene Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112021006844A priority Critical patent/BR112021006844A8/pt
Priority to DK19870299.5T priority patent/DK3864152T3/da
Priority to EA202191004A priority patent/EA202191004A1/ru
Priority to AU2019357450A priority patent/AU2019357450B2/en
Priority to CA3115864A priority patent/CA3115864A1/en
Priority to EP19870299.5A priority patent/EP3864152B1/en
Priority to EP23184286.5A priority patent/EP4306116A3/en
Priority to PE2021000498A priority patent/PE20211402A1/es
Priority to JP2021520227A priority patent/JP7252328B2/ja
Priority to CN202210724157.3A priority patent/CN116218836A/zh
Priority to US17/284,232 priority patent/US20210355494A1/en
Priority to SG11202103666PA priority patent/SG11202103666PA/en
Priority to ES19870299T priority patent/ES2962434T3/es
Priority to MX2021004187A priority patent/MX2021004187A/es
Priority to CN202210717093.4A priority patent/CN115651927B/zh
Priority to CR20210243A priority patent/CR20210243A/es
Priority to CN202210740517.9A priority patent/CN116042611A/zh
Priority to CN201980067380.1A priority patent/CN113631708B/zh
Application filed by Peking University, Edigene Inc. filed Critical Peking University
Priority to KR1020217014349A priority patent/KR102666695B1/ko
Publication of WO2020074001A1 publication Critical patent/WO2020074001A1/en
Priority to IL282191A priority patent/IL282191A/en
Priority to US17/226,961 priority patent/US20210310026A1/en
Priority to PH12021550805A priority patent/PH12021550805A1/en
Priority to CONC2021/0005956A priority patent/CO2021005956A2/es
Priority to ECSENADI202133637A priority patent/ECSP21033637A/es
Priority to JP2023045897A priority patent/JP2023078373A/ja
Priority to AU2023202184A priority patent/AU2023202184A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/712Nucleic acids or oligonucleotides having modified sugars, i.e. other than ribose or 2'-deoxyribose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/34Allele or polymorphism specific uses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/50Biochemical production, i.e. in a transformed host cell
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Definitions

  • the present invention is related to methods and compositions for editing RNAs using an engineered RNA capable of recruiting an adenosine deaminase to deaminate one or more adenosines in target RNAs.
  • Genome editing is a powerful tool for biomedical research and development of therapeutics for diseases. So far, the most popular gene editing technology is the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) -Cas system, which was developed from the adaptive immune system of bacteria and archaea. CRISPR-Cas can precisely target and cleave genome DNA, generating Double-Strand DNA Breaks (DSB) . DSB can be repaired through non-homologous end joining (NHEJ) pathways, resulting in an insertion or deletion (Indel) , which, in most cases, inactivates the gene. Alternatively, the homology-directed repair (HDR) pathway can repair the DSB using homologous templates dsDNA or ssDNA, and thus achieve precise genome editing.
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • ADAR Adenosine Deaminase Acting on RNA
  • Adar1 two isoforms, p110 and p150
  • Adar2 Adar3
  • the catalytic substrate of ADAR protein is double-stranded RNA, and it can remove the-NH 2 group from an adenosine (A) nucleobase, changing A to inosine (I) , which is recognized as guanosine (G) and paired with cytidine (C) during subsequent cellular transcription and translation processes.
  • A adenosine
  • I inosine
  • G guanosine
  • C cytidine
  • RNA editing include fusing antisense RNA to R/G motif (ADAR-recruiting RNA scaffold) to edit target RNA by overexpressing Adar1 or Adar2 proteins in mammalian cells, and using dCas13-ADAR to precisely target and edit RNA.
  • Nucleic acid editing carries enormous potential for biological research and the development of therapeutics.
  • Current tools for DNA or RNA editing rely on introducing exogenous proteins into living organisms, which is subject to potential risks or technical barriers due to possible aberrant effector activity, delivery limits and immunogenicity.
  • the present application provides a programmable approach that employs a short RNA to leverage endogenous ADAR (Adenosine Deaminase Acting on RNA) proteins for targeted RNA editing.
  • ADAR Addenosine Deaminase Acting on RNA
  • the present application provides an engineered RNA that is partially complementary to the target transcript to recruit native ADAR1 or ADAR2 to change adenosine to inosine at a specific site in a target RNA.
  • L everaging E ndogenous A DAR for P rogrammable E diting on R NA L everaging E ndogenous A DAR for P rogrammable E diting on R NA
  • ADAR-recruiting RNAs are referred to interchangeably as “dRNA” or “arRNA. ”
  • the present application provides a method for editing on a target RNA in a host cell, comprising introducing a deaminase-recruiting RNA (dRNA) or a construct encoding the deaminase-recruiting RNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an adenosine deaminase acting on RNA (ADAR) to deaminate a target adenosine (A) in the target RNA,.
  • the host cell is a eukaryotic cell.
  • the host cell is a mammalian cell.
  • the host cell is a human cell.
  • the host cell is a mouse cell.
  • the host cell is a prokaryotic cell.
  • the ADAR is naturally or endogenously present in the host cell, for example, naturally or endogenously present in the eukaryotic cell. In some embodiments, the ADAR is endogenously expressed by the host cell. In certain embodiments, the ADAR is exogenous to the host cell. In some embodiments, the ADAR is encoded by a nucleic acid (e.g., DNA or RNA) . In some embodiments, the method comprises introducing the ADAR or a construct encoding the ADAR into the host cell. In some embodiments, the method does not comprise introducing any protein into the host cell. In certain embodiments, the ADAR is ADAR1 and/or ADAR 2. In some embodiments, the ADAR is one or more ADARs selected from the group consisting of hADAR1, hADAR2, murine ADAR1 and murine ADAR2.
  • the dRNA is not recognized by a Cas. In some embodiments, the dRNA does not comprise crRNA, tracrRNA or gRNA used in a CRISPR/Cas system. In some embodiments, the method does not comprise introducing a Cas or Cas fusion protein into the host cell.
  • the deamination of the target A in the target RNA results in a missense mutation, an early stop codon, aberrant splicing, or alternative splicing in the target RNA.
  • the target RNA encodes a protein, and the deamination of the target A in the target RNA results in a point mutation, truncation, elongation and/or misfolding of the protein.
  • the deamination of the target A in the target RNA results in reversal of a missense mutation, an early stop codon, aberrant splicing, or alternative splicing in the target RNA.
  • the deamination of the target A in the target RNA results in a functional, full-length, correctly-folded and/or wild-type protein by reversal of a missense mutation, an early stop codon, aberrant splicing, or alternative splicing in the target RNA.
  • the target RNA is a regulatory RNA, and the deamination of the target A results in change in the expression of a downstream molecule regulated by the target RNA.
  • the method is for leveraging an endogenous adenosine deaminase for editing on a target RNA to generate point mutation and/or misfolding of the protein encoded by the target RNA, and/or generating an early stop codon, an aberrant splice site, and/or an alternative splice site in the target RNA.
  • a method for editing a plurality of target RNAs in host cells comprising introducing a plurality of dRNAs or constructs encoding the a plurality of dRNAs into the host cells, wherein each of the plurality of deaminase-recruiting RNAs comprises a complementary RNA sequence that hybridizes to a corresponding target RNA in the plurality of target RNAs, and wherein each dRNA is capable of recruiting an adenosine deaminase acting on RNA (ADAR) to deaminate a target adenosine (A) in the corresponding target RNA.
  • ADAR adenosine deaminase acting on RNA
  • an edited RNA or a host cell having an edited RNA produced by any one of the methods of RNA editing as described above.
  • the present application provides a method for treating or preventing a disease or condition in an individual, comprising editing a target RNA associated with the disease or condition in a cell of the individual according to any one of the methods for RNA editing as described above.
  • the method comprises editing the target RNA in the cell ex vivo.
  • the method comprises administering the edited cell to the individual.
  • the method comprises administering to the individual an effective amount of the dRNA or construct encoding the dRNA.
  • the method further comprises introducing to the cell the ADAR or a construct (e.g., viral vector) encoding the ADAR.
  • the method further comprises administering to the individual the ADAR or a construct (e.g., viral vector) encoding the ADAR.
  • the disease or condition is a hereditary genetic disease.
  • the disease or condition is associated with one or more acquired genetic mutations, e.g., drug resistance.
  • dRNA for deamination of a target adenosine in a target RNA by recruiting an Adenosine Deaminase Acting on RNA (ADAR) , comprising a complementary RNA sequence that hybridizes to the target RNA.
  • ADAR Adenosine Deaminase Acting on RNA
  • the dRNA comprises a nucleic acid sequence of any one of SEQ ID NOs: 25-44, 142-205, 341-342.
  • the dRNA comprises a RNA sequence comprising a cytidine (C) , adenosine (A) or uridine (U) directly opposite the target adenosine to be edited in the target RNA.
  • the RNA sequence further comprises one or more guanosines each directly opposite a non-target adenosine (s) in the target RNA.
  • the 5’ nearest neighbor of the target A in the target RNA sequence is a nucleotide selected from U, C, A and G with the preference U>C ⁇ A> G and the 3’ nearest neighbor of the target A in the target RNA sequence is a nucleotide selected from G, C, A and U with the preference G>C>A ⁇ U.
  • the target A is in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA.
  • the dRNA comprises an A directly opposite the U in the three-base motif, a C directly opposite the target A, and a C, G or U directly opposite the G in the three-base motif.
  • the dRNA comprises ACC, ACG or ACU opposite the UAG of the target RNA.
  • the deaminase-recruiting RNA comprises more than 40, 45, 50, 55, 60, 65, 70, 75 or 80 nucleotides.
  • the deaminase-recruiting RNA is 40-260, 45-250, 50-240, 60-230, 65-220, 70-210, 70-200, 70-190, 70-180, 70-170, 70-160, 70-150, 70-140, 70-130, 70-120, 70-110, 70-100, 70-90, 70-80, 75-200, 80-190, 85-180, 90-170, 95-160, 100-150 or 105-140 nucleotides in length.
  • the target RNA is a RNA selected from the group consisting of a pre-messenger RNA, a messenger RNA, a ribosomal RNA, a transfer RNA, a long non-coding RNA and a small RNA (e.g., miRNA) .
  • the dRNA is a single-stranded RNA.
  • the complementary RNA sequence is single-stranded, and wherein the dRNA further comprises one or more double-stranded regions.
  • the dRNA comprises one or more modifications, such as 2’ -O-methyl modification and/or phosphorothioate modification.
  • a construct encoding any one of the dRNA described above.
  • the construct comprises a promoter operably linked to a sequence encoding the dRNA.
  • the construct is a DNA construct.
  • the construct is a viral vector, such as an AAV, such as AAV8.
  • a library comprising a plurality of the dRNAs according to any one of the dRNAs described above or a plurality of the constructs according to any one of the constructs described above.
  • compositions, host cells, kits and articles of manufacture comprising any one of the dRNAs described herein, any one of the constructs described herein, or any one of the libraries described herein.
  • FIGs. 1A-1H show RNA editing with single dRNA utilizing endogenous Adar1 protein.
  • FIG. 1A and 1B show schematic representations of RNA editing with endogenous Adar1 protein.
  • FIG. 1C shows editing reporter mRNA with dRNA using endogenous Adar1 protein.
  • FIG. 1D shows statistical analysis of the results in FIG. 1B.
  • FIG. 1E shows Adar1 knockout and Adar1 (p110) , Adar1 (p150) and Adar2 rescue results.
  • FIG. 1F shows statistical analysis of the results in FIG. 1D.
  • FIG. 1G shows the effect of Adar1 (p110) , Adar1 (p150) or Adar2 overexpression on RNA editing mediated by dRNA in 293T-WT cells.
  • FIG. 1H shows that deep sequencing (i.e., Next Generation Sequencing, NGS) results confirmed A to G editing in the targeting site.
  • deep sequencing i.e., Next Generation Sequencing, NGS
  • FIGs. 2A-2H show optimization of dRNAs.
  • FIG. 2A shows schematic representation of four kinds of base (A, U, C and G) identify opposite to the targeting adenosine.
  • FIG. 2B shows effects of base identify opposite to the targeting adenosine on RNA editing efficiency by dRNA.
  • FIG. 2C shows schematic representation of dRNA with one, two or three bases mismatched with UAG targeting site.
  • FIG. 2D shows effects of one, two or three bases mismatched with UAG targeting site on Reporter RNA editing by dRNA.
  • dRNA preferred A-C mismatch on the targeting adenosine.
  • FIG. 2E shows schematic representation of dRNA with variant length.
  • FIG. 2F shows the effect of dRNA length on RNA editing efficiency based on dual fluorescence reporter-2.
  • FIG. 2G shows schematic representation of different A-C mismatch position.
  • FIG. 2H shows effect of A-C mismatch position on RNA editing efficiency.
  • FIGs. 3A-3B show editing flexibility for endogenous RNA editing through exemplary RNA editing method of the present application.
  • FIG. 3A shows percentage quantification of endogenous RNA editing efficiency at all 16 different 3-base motifs.
  • FIG. 3B shows heatmap of 5’ and 3’ base preferences of endogenous RNA editing for 16 different 3 base motifs.
  • FIGs. 4A-4H show editing the mRNA of endogenous genes with dRNA in 293T cells.
  • FIG. 4A shows schematic representation of KRAS mRNA target and dRNA with variant length.
  • FIG. 4B shows editing the mRNA of endogenous KRAS gene with dRNA in 293T cells. Empty vector, dRNA-91nt plasmids were transfected into 293T-WT cells, respectively. 60 hours later, the RNA was isolated for RT-PCR, and then cDNA was amplified and sequenced on Illumina NextSeq.
  • FIG. 4C shows schematic representation of PPIB mRNA target (site1, site2 and site3) and the corresponding dRNA design.
  • FIG. 4D, 4E and 4F show editing the mRNA of endogenous PPIB gene with dRNA in 293T cells.
  • FIG. 4G shows schematic representation of ⁇ -Actin mRNA target and dRNA (71-nt and 131-nt) .
  • FIG. 4H shows editing the mRNA of endogenous ⁇ -Actin gene with dRNA in 293T cells.
  • FIGs. 5A-5G show off-target analysis.
  • FIG. 5A shows schematic representation of the sequence window in which A to I edits were analyzed for PPIB mRNA target (PPIB site 1) .
  • the black arrow indicates the targeted adenosine.
  • FIG. 5B shows deep sequencing quantification of A to I RNA editing by 151-nt dRNA targeting PPIB mRNA target (PPIB site 1) .
  • FIG. 5C shows schematic representation of the sequence window in which A to I edits were analyzed for KRAS mRNA target.
  • the black arrow indicates the targeted adenosine.
  • FIG. 5D shows deep sequencing quantification of A to I RNA editing by 91-nt and 111-nt dRNA targeting KRAS mRNA target.
  • FIG. 5A shows schematic representation of the sequence window in which A to I edits were analyzed for PPIB mRNA target (PPIB site 1) .
  • the black arrow indicates the targeted adenos
  • FIG. 5E shows schematic representation of designed four kinds of 91-nt or 111-nt dRNA variants containing different A-G mismatch combinations.
  • the A-G mismatch was designed based on the statistical results in FIG. 5D and existing knowledge on genic codes for different amino acids.
  • FIG. 5F shows the results of targeted A56 editing by dRNA and different kinds of dRNA variants in FIG. 5E.
  • FIG. 5G shows deep sequencing quantification of A to I RNA editing by 111-nt dRNA and four kinds of 111-nt dRNA variants targeting KRAS mRNA target.
  • FIGs. 6A-6H show RNA editing with single dRNA utilizing endogenous Adar1 protein.
  • FIG. 6A shows schematic representation of RNA editing by dLbuCas13-ADARDD fusion proteins. The catalytically inactive dLbuCas13 was fused to the RNA deaminase domains of ADAR1 or ADAR2.
  • FIG. 6B shows schematic representation of dual fluorescence reporter mRNA target and guide RNA design.
  • FIG. 6C shows statistical analysis of the results in FIGs. 6A and 6B.
  • FIG. 6D shows the mRNA level of Adar1 and Adar2 in 293T-WT cells.
  • FIG. 6E shows genotyping results of ADAR1 gene in 293T-ADAR1-KO cell lines by genome PCR.
  • FIG. 6F shows the expression level of Adar1 (p110) and Adar1 (p150) in 293T-WT and 293T-ADAR1-KO cell lines via western blotting.
  • FIG. 6G shows the effects of Adar1 (p110) , Adar1 (p150) or Adar2 overexpression on RNA editing mediated by dRNA in 293T-WT cells via FACS.
  • FIG. 6H shows Sanger sequencing results showed A to G editing in the targeted adenosine site.
  • FIGs. 7A-7C shows optimization of dRNAs.
  • FIG. 7A shows schematic representation of dRNA with variant length and the targeted mRNA editing results by dRNA with variant length based on dual fluorescence reporter-1.
  • FIG. 7B shows schematic representation of different A-C mismatch position and the effect of A-C mismatch position on RNA editing efficiency based on dual fluorescence reporter-1.
  • FIG. 7C shows schematic representation of different A-C mismatch position and the effect of A-C mismatch position on RNA editing efficiency based on dual fluorescence reporter-3.
  • FIGs. 8A-8B shows editing the mRNA of endogenous genes with dRNA in 293T cells.
  • FIG. 8A shows editing the mRNA of endogenous ⁇ -Actin gene (site2) with dRNA in 293T cells.
  • FIG. 8B shows editing the mRNA of endogenous GAPDH gene with dRNA in 293T cells.
  • FIG. 9 shows RNA editing by dRNA in different cell lines.
  • FIG. 9A shows that reporter plasmids and dRNA plasmids were co-transfected into different cell lines, and the results showed that dRNA could function well in multiple cell lines, indicating the universality of dRNA application.
  • FIGs. 10A-10D show exploration of an efficient exemplary RNA editing platform.
  • FIG. 10A Schematic of dLbuCas13a-ADAR1 DD (E1008Q) fusion protein and the corresponding crRNA.
  • the catalytic inactive LbuCas13a was fused to the deaminase domain of ADAR1 (hyperactive E1008Q variant) using 3 ⁇ GGGGS linker.
  • the crRNA (crRNA Cas13a ) consisted of Lbu-crRNA scaffold and a spacer, which was complementary to the targeting RNA with an A-C mismatch as indicated.
  • FIG. 10B Schematic of dual fluorescent reporter system and the Lbu-crRNA with various lengths of spacers as indicated.
  • FIG. 10A Schematic of dLbuCas13a-ADAR1 DD (E1008Q) fusion protein and the corresponding crRNA.
  • the catalytic inactive LbuCas13a was fused to the deamina
  • FIG. 10C Quantification of the EGFP positive (EGFP + ) cells.
  • FIG. 10D Representative FACS result from the experiment performed with the control (Ctrl crRNA 70 ) or the targeting spacer (crRNA 70 ) .
  • FIGs. 11A-11G show exemplary methods of leveraging endogenous ADAR1 protein for targeted RNA editing.
  • FIG. 11A Schematic of the Reporter-1 and the 70-nt arRNA.
  • FIG. 11B Representative FACS analysis of arRNA-induced EGFP expression in wild-type (HEK293T, upper) or ADAR1 knockout (HEK293T ADAR1 –/– , lower) cells stably expressing the Repoter-1.
  • FIG. 11C Western blot analysis showing expression levels of ADAR1 proteins in wild-type and HEK293T ADAR1 –/– cells, as well as those in HEK293T ADAR1 –/– cells transfected with ADAR1 isoforms (p110 and p150) .
  • FIG. 11D Western blot analysis showing expression levels of ADAR2 proteins in wild-type and HEK293T ADAR1 –/– cells, as well as those in HEK293T ADAR1 –/– cells transfected with ADAR2.
  • FIG. 11E Quantification of the EGFP positive (EGFP + ) cells. Reporter-1 and indicated ADAR-expressing constructs were co-transfected into HEK293T ADAR1 –/– cells, along with the Ctrl RNA 70 or with the
  • FIG. 11F The Electropherograms showing Sanger sequencing results in the Ctrl RNA 70 (upper) or the arRNA 70 (lower) -targeted region.
  • FIG. 11G Quantification of the A to I conversion rate at the targeted site by deep sequencing.
  • FIGs. 12A-12B show mRNA expression level of ADAR1/ADAR2 and arRNA-mediated RNA editing.
  • FIG. 12B Representative FACS results from Fig. 1E.
  • FIGs. 14A-14D show targeted RNA editing with an exemplary LEAPER method in multiple cell lines.
  • FIG. 14A Western-blot results showing the expression levels of ADAR1, ADAR2 and ADAR3 in indicated human cell lines. ⁇ -tubulin was used as a loading control. Data shown is the representative of three independent experiments.
  • ADAR1 –/– /ADAR2 represents ADAR1-knockout HEK293T cells overexpressing ADAR2.
  • FIG. 14B Relative ADAR protein expression levels normalized by ⁇ -tubulin expression.
  • FIG. 14A Western-blot results showing the expression levels of ADAR1, ADAR2 and ADAR3 in indicated human cell lines. ⁇ -tubulin was used as a loading control. Data shown is the representative of three independent experiments.
  • ADAR1 –/– /ADAR2 represents ADAR1-knockout HEK293T cells overexpressing ADAR2.
  • FIG. 14B Relative ADAR protein expression levels normalized by ⁇ -tubulin expression.
  • FIG. 14C Indicated human cells were transfected with Reporter-1, along with the 71-nt control arRNA (Ctrl RNA 71 ) or with the 71-nt targeting arRNA (arRNA 71 ) followed by FACS analysis.
  • FIGs. 15A-15C show schematics of Reporter-1 (a) , -2 (b) , and-3 (c) , as well as their corresponding arRNAs.
  • FIGs. 16A-16G show characterization and optimization of exemplary LEAPER methods.
  • FIG. 16A Top, schematic of the design of arRNAs with changed triplet (5’ -CNA, N denotes A, U, C or G) opposite to the target UAG. Bottom, EGFP + percent showing the effects of variable bases opposite to the targeted adenosine on RNA editing efficiency.
  • FIG. 16B Top, the design of arRNAs with changed neighboring bases flanking the cytidine in the A-C mismatch (5’ -N 1 CN 2 ) . Bottom, the effects of 16 different combinations of N 1 CN 2 on RNA editing efficiency.
  • FIG. 16A Top, schematic of the design of arRNAs with changed triplet (5’ -CNA, N denotes A, U, C or G) opposite to the target UAG. Bottom, EGFP + percent showing the effects of variable bases opposite to the targeted adenosine on RNA editing efficiency.
  • FIG. 16B Top, the design of arRNAs with changed neighbor
  • FIG. 16C Summary of the preference of 5’ and 3’ nearest neighboring sites of the cytidine in the A-C mismatch.
  • FIG. 16D Top, the design of arRNAs with variable length. Bottom, the effect of arRNA length on RNA editing efficiency based on Reporter-1 and Reporter-2.
  • FIG. 16E Top, the design of arRNAs with variable A-C mismatch position. Bottom, the effect of A-C mismatch position on RNA editing efficiency based on Reporter 1 and Reporter-2.
  • FIG. 16F Top, the design of the triplet motifs in the reporter-3 with variable nearest neighboring bases surrounding the targeting adenosine (5’ -N 1 AN 2 ) and the opposite motif (5’ -N 2 CN 1 ) on the 111-nt arRNA (arRNA 111 ) .
  • FIGs. 17A-17I show editing of endogenous transcripts with exemplary LEAPER methods.
  • FIG. 17A Schematic of the targeting endogenous transcripts of four disease-related genes (PPIB, KRAS, SMAD4 and FANCC) and the corresponding arRNAs.
  • FIG. 17B Deep sequencing results showing the editing rate on targeted adenosine of the PPIB, KRAS, SMAD4 and FANCC transcripts by introducing indicated lengths of arRNAs.
  • FIG. 17C Deep sequencing results showing the editing rate on non-UAN sites of endogenous PPIB, FANCC and IDUA transcripts.
  • FIG. 17D Multiplex editing rate by two 111-nt arRNAs.
  • FIG. 17E Schematic of the PPIB transcript sequence covered by the 151-nt arRNA. The black arrow indicates the targeted adenosine. All adenosines were marked in red.
  • FIG. 17F Heatmap of editing rate on adenosines covered by indicated lengths of arRNAs targeting the PPIB gene (marked in bold frame in blue) .
  • FIG. 17G Top, the design of the triplet motifs in the reporter-3 with variable nearest neighboring bases surrounding the targeting adenosine (5’ -N 1 AN 2 ) and the opposite motif (5’ -N 2’ GN 1’ ) in the 111-nt arRNA (arRNA 111 ) .
  • Bottom deep sequencing results showing the editing rate.
  • FIG. 17H Top, the design of arRNAs with two consecutive mismatches in the 5’-N 1 GN 2 motif opposite to the 5’ -UAG or the 5’ -AAG motifs. Deep sequencing results showing the editing rate by an arRNA 111 with two consecutive mismatches in the 5’ -N 1 GN 2 motif opposite to the 5’ -UAG motif (bottom left) or the 5’ -AAG motif (bottom right) .
  • FIG. 17I Heatmap of the editing rate on adenosines covered by engineered arRNA 111 variants targeting the KRAS gene. Data in FIGs. 17B, 17C, 17D, 17G and 17H are presented as the mean ⁇ s.e.m.
  • FIGs. 18A-18B show effects of exemplary LEAPER methods on the expression levels of targeted transcripts and protein products.
  • FIG. 18B Western blot results showing the effects on protein products of targeted KRAS gene by 151-nt arRNA in HEK293T cells. ⁇ -tubulin was used as a loading control.
  • FIGs. 19A-19F show editing of endogenous transcripts with exemplary LEAPER methods.
  • FIG. 19A Schematic of the KARS transcript sequence covered by the 151-nt arRNA. The arrow indicates the targeting adenosine. All adenosines were marked in red.
  • FIG. 19B Heatmap of editing rate on adenosines covered by indicated arRNAs in the KARS transcript (marked in the bold frame in blue) .
  • FIG. 19C Schematic of the SMAD4 transcript covered by the 151-nt arRNA.
  • FIG. 19D Heatmap of editing rate on adenosines covered by indicated arRNAs in the SMAD4 transcript.
  • FIG. 19E Schematic of the FANCC transcript covered by the 151-nt arRNA.
  • FIGs. 20A-20D show transcriptome-wide specificity of RNA editing by LEAPER.
  • FIGs. 20A and 20B Transcriptome-wide off-targeting analysis of Ctrl RNA 151 and arRNA 151 -PPIB. The on-targeting site (PPIB) is highlighted in red. The potential off-target sites identified in both Ctrl RNA and PPIB-targeting RNA groups are labeled in blue.
  • FIG. 20C The predicted annealing affinity between off-target sites and the corresponding Ctrl RNA 151 or arRNA 151 -
  • FIG. 20D Top, schematic of the highly complementary region between arRNA 151 -PPIB and the indicated potential off-target sites, which were predicted by searching homologous sequences through NCBI-BLAST.
  • FIGs. 21A-21B show evaluation of potential off-targets.
  • FIG. 21A Schematic of the highly complementary region of arRNA 111 -FANCC and the indicated potential off-target sequence, which were predicted by searching homologous sequences through NCBI-BLAST.
  • FIGs. 22A-22F show safety evaluation of applying exemplary LEAPER methods in mammalian cells.
  • FIGs. 22A and 22B Transcriptome-wide analysis of the effects of Ctrl RNA 151 (a) arRNA 151 -PPIB (b) on native editing sites by transcriptome-wide RNA-sequencing. Pearson’s correlation coefficient analysis was used to assess the differential RNA editing rate on native editing sites.
  • FIGs. 22C and 22D Differential gene expression analysis of the effects of Ctrl RNA 151 (c) arRNA 151 -PPIB (d) with RNA-seq data at the transcriptome level. Pearson’s correlation coefficient analysis was used to assess the differential gene expression.
  • FIGs. 22A and 22B Transcriptome-wide analysis of the effects of Ctrl RNA 151 (a) arRNA 151 -PPIB (b) on native editing sites by transcriptome-wide RNA-sequencing. Pearson’s correlation coefficient analysis was used to assess the differential RNA editing rate
  • FIGs. 23A-23D show recovery of transcriptional regulatory activity of mutant TP53W53X by LEAPER.
  • FIG. 23A Top, Schematic of the TP53 transcript sequence covered by the 111-nt arRNA containing c. 158G>A clinical-relevant non-sense mutation (Trp53Ter) .
  • the black arrow indicates the targeted adenosine. All adenosines were marked in red.
  • FIG. 23B Deep sequencing results showing the targeted editing on TP53 W53X transcripts by arRNA 111 , arRNA 111 -AG1 and arRNA 111 -AG4.
  • FIG. 23C Western blot showing the recovered production of full-length p53 protein from the TP53 W53X transcripts in the HEK293T TP53 –/– cells.
  • FIG. 24 show editing of mutant TP53W53X transcripts by an exemplary LEAPER method.
  • Top schematic of the TP53 transcript sequence covered by the 111-nt arRNAs. The arrow indicates the targeted adenosine. All adenosines were marked in red.
  • Bottom a heatmap of editing rate on adenosines covered by indicated arRNAs in the TP53 transcript.
  • FIG. 25 shows a schematic representation of the selected disease-relevant cDNA containing G to A mutation from ClinVar data and the corresponding 111-nt arRNA.
  • FIG. 26 shows correction of pathogenic mutations by an exemplary LEAPER method.
  • FIGs. 27A-27C show RNA editing in multiple human primary cells by exemplary LEAPER methods.
  • FIG. 27A Quantification of the EGFP positive (EGFP + ) cells induced by LEAPER-mediated RNA editing.
  • Human primary pulmonary fibroblasts and human primary bronchial epithelial cells were transfected with Reporter-1, along with the 151-nt control RNA (Ctrl RNA 151 ) or the 151-nt targeting arRNA (arRNA 151 ) followed by FACS analysis.
  • FIGs. 27B and 27C Deep sequencing results showing the editing rate on PPIB transcripts in human primary pulmonary fibroblasts, human primary bronchial epithelial cells (b) , and human primary T cells (c) .
  • FIGs. 28A-28D show targeted editing by lentiviral transduction of arRNA and electroporation of synthesized arRNA oligonucleotides.
  • FIG. 28A Quantification of the EGFP + cells. HEK293T cells stably expressing the Repoter-1 were infected with lentivirus expressing 151-nt of Ctrl RNA or the targeting arRNA. FACS analyses were performed 2 days and 8 days post infection. The ratios of EGFP + cells were normalized by lentiviral transduction efficiency (BFP + ratios) .
  • FIG. 28B Deep sequencing results showing the editing rate on the PPIB transcripts upon lentiviral transduction of 151-nt arRNAs into HEK293T cells.
  • FIG. 28A Quantification of the EGFP + cells.
  • HEK293T cells stably expressing the Repoter-1 were infected with lentivirus expressing 151-nt of Ctrl RNA or the targeting arRNA.
  • FIG. 28C Schematic of the PPIB sequence and the corresponding 111-nt targeting arRNA. * (in red) represents nucleotide with 2’ -O-methyl and phosphorothioate linkage modifications.
  • FIG. 28D Deep sequencing results showing the editing rate on the PPIB transcripts upon electroporation of 111-nt synthetic arRNA oligonucleotides into human primary T cells.
  • FIGs. 29A-29E show restoration of ⁇ -L-iduronidase activity in Hurler syndrome patient-derived primary fibroblast by an exemplary LEAPER method.
  • FIG. 29A Top, genetic information of pathogenic mutation in patient-derived fibroblast GM06214; Medium, schematic of the IDUA mature mRNA sequence of GM06214 cells (Black) containing a homozygous TGG>TAG mutation in exon 9 of the IDUA gene (Trp402Ter) , and the corresponding 111-nt targeting arRNA 111 -IDUA-V1 (Blue) ; Bottom, schematic of the IDUA pre-mRNA sequence of GM06214 cells (Black) and the corresponding 111-nt targeting arRNA 111 -IDUA-V2 (Blue) .
  • FIG. 29C Deep sequencing results showing the targeted editing rate on IDUA transcripts in GM06214 cells, 48 hours post electroporation.
  • FIG. 29D Top, schematic of the IDUA transcript sequence covered by the 111-nt arRNAs. The arrow indicates the targeted adenosine. All adenosines were marked in red.
  • FIGs. 30A-30C shows three versions of dual fluorescence reporters (Reporter-1, -2 and-3) , mCherry and EGFP.
  • FIG. 30A structure of Reporter-1
  • FIG. 30B structure of Reporter-2
  • FIG. 30C structure of Reporter-3.
  • FIG. 31 shows the plasmid information of AAV8-IDUA-adRNA151-KD2 and AAV8-Random151-KD2.
  • FIG. 32A-B show ⁇ -L-iduronidase relative enzymatic activity of MPSI mice injected with AAV8-IDUA-adRNA151-KD2 and AAV8-Random151-KD2. GM01323 cells as a control.
  • FIG. 33A-B show the RNA editing efficiency of MPSI mice injected with AAV8-IDUA-adRNA151-KD2 and AAV8-Random151-KD2.
  • RNA editing methods referred herein as “LEAPER” methods
  • dRNAs deaminase-recruiting RNAs
  • arRNAs ADAR-recruiting RNAs
  • the dRNA acts through hybridizing to its target RNA in a sequence-specific fashion to form a double-stranded RNA, which recruits an Adenosine Deaminase Acting on RNA (ADAR) to deaminate a target adenosine in the target RNA.
  • ADAR Adenosine Deaminase Acting on RNA
  • efficient RNA editing can be achieved in some embodiments without ectopic or overexpression of the ADAR proteins in the host cell.
  • methods and compositions for treating or preventing a disease or condition in an individual using the RNA editing methods are also provided.
  • RNA editing methods described herein do not use fusion proteins comprising an ADAR and a protein that specifically binds to a guide nucleic acid, such as Cas.
  • the deaminase-recruiting RNAs ( “dRNA” ) described herein do not comprise crRNA, tracrRNA or gRNA used in the CRISPR/Cas system.
  • the dRNA does not comprise an ADAR-recruiting domain, or chemical modification (s) .
  • the dRNA can be expressed from a plasmid or a viral vector, or synthesized as an oligonucleotide, which could achieve desirable editing efficiency.
  • the LEAPER methods described herein have manageable off-target rates on the targeted transcripts and rare global off-targets. Inventors have used the LEAPER method to restore p53 function by repairing a specific cancer-relevant point mutation.
  • the LEAPER methods described herein can also be applied to a broad spectrum of cell types including multiple human primary cells, and can be used to restore the ⁇ -L-iduronidase catalytic activity in Hurler syndrome patient-derived primary fibroblasts without evoking innate immune responses.
  • the LEAPER method involves a single molecule (i.e., dRNA) system.
  • dRNA single molecule
  • RNA deaminase-recruiting RNA, ” “dRNA, ” “ADAR-recruiting RNA” and “arRNA” are used herein interchangeably to refer to an engineered RNA capable of recruiting an ADAR to deaminate a target adenosine in an RNA.
  • nucleotide refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • nucleobases refer to the nucleobases as such.
  • adenosine refers to the nucleobases linked to the ribose or deoxyribose sugar moiety.
  • nucleoside refers to the nucleobase linked to the ribose or deoxyribose.
  • nucleotide refers to the respective nucleobase-ribosyl-phosphate or nucleobase-deoxyribosyl-phosphate.
  • adenosine and adenine with the abbreviation, “A” )
  • guanosine and guanine with the abbreviation, “G”
  • cytosine and cytidine with the abbreviation, “C”
  • uracil and uridine with the abbreviation, “U” )
  • thymine and thymidine with the abbreviation, “T”
  • inosine and hypo-xanthine with the abbreviation, “I”
  • nucleobase, nucleoside and nucleotide are used interchangeably, unless the context clearly requires differently.
  • target RNA refers to an RNA sequence to which a deaminase-recruiting RNA sequence is designed to have perfect complementarity or substantial complementarity, and hybridization between the target sequence and the dRNA forms a double stranded RNA (dsRNA) region containing a target adenosine, which recruits an adenosine deaminase acting on RNA (ADAR) that deaminates the target adenosine.
  • dsRNA double stranded RNA
  • ADAR adenosine deaminase acting on RNA
  • the ADAR is naturally present in a host cell, such as a eukaryotic cell (preferably, a mammalian cell, more preferably, a human cell) .
  • the ADAR is introduced into the host cell.
  • complementarity refers to the ability of a nucleic acid to form hydrogen bond (s) with another nucleic acid by traditional Watson-Crick base-pairing.
  • a percent complementarity indicates the percentage of residues in a nucleic acid molecule which can form hydrogen bonds (i.e., Watson-Crick base pairing) with a second nucleic acid (e.g., about 5, 6, 7, 8, 9, 10 out of 10, being about 50%, 60%, 70%, 80%, 90%, and 100%complementary respectively) .
  • Perfectly complementary means that all the contiguous residues of a nucleic acid sequence form hydrogen bonds with the same number of contiguous residues in a second nucleic acid sequence.
  • substantially complementary refers to a degree of complementarity that is at least about any one of 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 100%over a region of about 40, 50, 60, 70, 80, 100, 150, 200, 250 or more nucleotides, or refers to two nucleic acids that hybridize under stringent conditions.
  • stringent conditions for hybridization refer to conditions under which a nucleic acid having complementarity to a target sequence predominantly hybridizes with the target sequence, and substantially does not hybridize to non-target sequences. Stringent conditions are generally sequence-dependent, and vary depending on a number of factors. In general, the longer the sequence, the higher the temperature at which the sequence specifically hybridizes to its target sequence. Non-limiting examples of stringent conditions are described in detail in Tijssen (1993) , Laboratory Techniques In Biochemistry And Molecular Biology-Hybridization With Nucleic Acid Probes Part I, Second Chapter “Overview of principles of hybridization and the strategy of nucleic acid probe assay” , Elsevier, N, Y.
  • Hybridization refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues.
  • the hydrogen bonding may occur by Watson Crick base pairing, Hoogstein binding, or in any other sequence specific manner.
  • a sequence capable of hybridizing with a given sequence is referred to as the "complement" of the given sequence.
  • the terms “cell” , “cell line” , and “cell culture” are used interchangeably and all such designations include progeny. It is understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as the original cells are included.
  • a method for editing a target RNA in a host cell comprising introducing a deaminase-recruiting RNA (dRNA) or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an adenosine deaminase acting on RNA (ADAR) to deaminate a target adenosine (A) in the target RNA.
  • a host cell e.g., eukaryotic cell
  • a method for editing a target RNA in a host cell comprising introducing a dRNA or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA recruits an endogenously expressed ADAR of the host cell to deaminate a target A in the target RNA.
  • the method does not comprise introducing any protein or construct encoding a protein (e.g., Cas, ADAR or a fusion protein of ADAR and Cas) to the host cell.
  • a method for editing a target RNA in a host cell comprising introducing: (a) a dRNA or a construct encoding the dRNA, and (b) an ADAR or a construct encoding the ADAR into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA recruits the ADAR to deaminate a target A in the target RNA.
  • the ADAR is an endogenously encoded ADAR of the host cell, wherein introduction of the ADAR comprises over-expressing the ADAR in the host cell.
  • the ADAR is exogenous to the host cell.
  • the construct encoding the ADAR is a vector, such as a plasmid, or a viral vector (e.g., a lentiviral vector) .
  • a method for editing a plurality (e.g., at least about 2, 3, 4, 5, 10, 20, 50, 100 or more) of target RNAs in a host cell comprising introducing a plurality of dRNAs or constructs encoding the plurality of dRNAs into the host cell, wherein each dRNA comprises a complementary RNA sequence that hybridizes to a corresponding target RNA in the plurality of target RNAs, and wherein each dRNA is capable of recruiting an ADAR to deaminate a target A in the corresponding target RNA.
  • a method for editing a plurality (e.g., at least about 2, 3, 4, 5, 10, 20, 50, 100 or more) of target RNAs in a host cell comprising introducing a plurality of dRNAs or constructs encoding the plurality of dRNAs into the host cell, wherein each dRNA comprises a complementary RNA sequence that hybridizes to a corresponding target RNA in the plurality of target RNAs, and wherein each dRNA recruits an endogenously expressed ADAR to deaminate a target A in the corresponding target RNA.
  • a method for editing a plurality (e.g., at least about 2, 3, 4, 5, 10, 20, 50, 100, 1000 or more) of target RNAs in a host cell comprising introducing: (a) a plurality of dRNAs or constructs encoding the plurality of dRNAs, and (b) an ADAR or a construct encoding ADAR into the host cell, wherein each dRNA comprises a complementary RNA sequence that hybridizes to a corresponding target RNA in the plurality of target RNAs, and wherein each dRNA recruits the ADAR to deaminate a target A in the corresponding target RNA.
  • a host cell e.g., a eukaryotic cell
  • the present application provides a method for editing a plurality of RNAs in host cells by introducing a plurality of the deaminase-recruiting RNAs, one or more constructs encoding the deaminase-recruiting RNAs, or a library described herein, into the host cells.
  • the method for editing on a target RNA comprises introducing multiple deaminase-recruiting RNAs or one or more constructs comprising the multiple deaminase-recruiting RNAs into host cells to recruit adenosine deaminase acting on RNA (ADAR) to perform deamination reaction on one or more target adenosines in one or more target RNAs, wherein each deaminase-recruiting RNA comprises a RNA sequences complementary to a corresponding target RNA.
  • ADAR adenosine deaminase acting on RNA
  • the present application provides a method for generating one or more modifications in a target RNA and/or the protein encoded by a target RNA in a host cell (e.g., eukaryotic cell) , comprising introducing a dRNA or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the one or more modifications are selected from the group consisting of a point mutation of the protein encoded by the target RNA, misfolding of the protein encoded by the target RNA, an early stop codon in the target RNA, an aberrant splice site in the target RNA, and an alternative splice site in the target RNA.
  • a host cell e.g., eukaryotic cell
  • the present application provides a method for restoring expression or activity of a target RNA or the protein encoded by a target RNA in a host cell (e.g., eukaryotic cell) , comprising introducing a dRNA or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, wherein the target RNA has G to A mutation that results in an early stop codon, an aberrant splice site, or an alternative splice site in the target RNA, or misfolding of the protein, and wherein the dRNA recruits a ADAR to edit the A mutation, thereby restoring expression or activity of the target RNA or the protein encoded by the target RNA.
  • a host cell e.g., eukaryotic cell
  • the method for generating one or more modifications in a plurality of target RNAs and/or the proteins encoded by the target RNAs in a host cell comprises introducing a plurality of deaminase-recruiting RNAs or constructs encoding the plurality of deaminase-recruiting RNAs into the host cell, wherein each dRNA comprises a complementary RNA sequence that hybridizes to a corresponding target RNA in the plurality of target RNAs, and wherein each dRNA is capable of recruiting an ADAR to deaminate a target A in the corresponding target RNA.
  • a host cell e.g., eukaryotic cell
  • the present application provides use of a deaminase-recruiting RNA according to any one of the dRNAs described herein for editing a target RNA in a host cell.
  • the deaminase-recruiting RNA comprises a complementary RNA sequence that hybridizes to the target RNA to be edited.
  • the present application provides use of a deaminase-recruiting RNA according to any one of the dRNAs described herein for generating one or more modifications on a target RNA and/or the protein encoded by a target RNA, wherein the one or more modifications are selected from a group consisting of a point mutation of the protein encoded by the target RNA, misfolding of the protein encoded by the target RNA, an early stop codon in the target RNA, an aberrant splice site in the target RNA, and an alternative splice site in the target RNA.
  • the deaminase-recruiting RNA comprises a complementary RNA sequence that hybridizes to the target RNA to be edited.
  • the invention also relates to a method for leveraging an endogenous adenosine deaminase for editing a target RNA in a eukaryotic cell, comprising introducing a dRNA or a construct encoding the dRNA, as described herein, into the eukaryotic cell to recruit naturally endogenous adenosine deaminase acting on RNA (ADAR) to perform deamination reaction on a target adenosine in the target RNA sequence.
  • ADAR naturally endogenous adenosine deaminase acting on RNA
  • the dRNA comprises more than about any one of 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nucleotides.
  • the dRNA is about any one of 40-260, 45-250, 50-240, 60-230, 65-220, 70-220, 70-210, 70-200, 70-190, 70-180, 70-170, 70-160, 70-150, 70-140, 70-130, 70-120, 70-110, 70-100, 70-90, 70-80, 75-200, 80-190, 85-180, 90-170, 95-160, 100-200, 100-150, 100-175, 110-200, 110-175, 110-150, or 105-140 nucleotides in length.
  • the dRNA is about 71 nucleotides long. In some embodiments, the dRNA is about 111 nucleotides long.
  • the dRNA does not comprise an ADAR-recruiting domain.
  • ADAR-recruiting domain can be a nucleotide sequence or structure that binds at high affinity to ADAR, or a nucleotide sequence that binds to a binding partner fused to ADAR in an engineered ADAR construct.
  • ADAR-recruiting domains include, but are not limited to, GluR-2, GluR-B (R/G) , GluR-B (Q/R) , GluR-6 (R/G) , 5HT2C, and FlnA (Q/R) domain; see, for example, Wahlstedt, Helene, and Marie, "Site-selective versus promiscuous A-to-I editing. " Wiley Interdisciplinary Reviews: RNA 2.6 (2011) : 761-771, which is incorporated herein by reference in its entirety.
  • the dRNA does not comprise a double-stranded portion.
  • the dRNA does not comprise a hairpin, such as MS2 stem loop.
  • the dRNA is single stranded.
  • the ADAR not comprise a DSB-binding domain.
  • the dRNA consists of (or consists essentially of) the complementary RNA sequence.
  • the dRNA does not comprise chemical modifications.
  • the dRNA does not comprise a chemically modified nucleotide, such as 2’ -O-methyl nucleotide or a nucleotide having a phosphorothioate linkage.
  • the dRNA comprises 2’ -O-methyl and phosphorothioate linkage modifications only at the first three and last three residues.
  • the dRNA is not an antisense oligonucleotide (ASO) .
  • ASO antisense oligonucleotide
  • the host cell is a prokaryotic cell. In some embodiments, the host cell is a eukaryotic cell. In some embodiments, the host cell is a mammalian cell. In some embodiments, the host cell is a human cell. In some embodiments, the host cell is a murine cell. In some embodiments, the host cell is a plant cell or a fungal cell.
  • the host cell is a cell line, such as HEK293T, HT29, A549, HepG2, RD, SF268, SW13 and HeLa cell.
  • the host cell is a primary cell, such as fibroblast, epithelial, or immune cell.
  • the host cell is a T cell.
  • the host cell is a post-mitosis cell.
  • the host cell is a cell of the central nervous system (CNS) , such as a brain cell, e.g., a cerebellum cell.
  • CNS central nervous system
  • a method of editing a target RNA in a primary host cell comprising introducing a dRNA or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA recruits an endogenously expressed ADAR of the host cell to deaminate a target A in the target RNA.
  • the ADAR is endogenous to the host cell.
  • the adenosine deaminase acting on RNA (ADAR) is naturally or endogenously present in the host cell, for example, naturally or endogenously present in the eukaryotic cell.
  • the ADAR is endogenously expressed by the host cell.
  • the ADAR is exogenously introduced into the host cell.
  • the ADAR is ADAR1 and/or ADAR2.
  • the ADAR is one or more ADARs selected from the group consisting of hADAR1, hADAR2, mouse ADAR1 and ADAR2.
  • the ADAR is ADAR1, such as p110 isoform of ADAR1 ( “ADAR1 p110 ” ) and/or p150 isoform of ADAR1 ( “ADAR1 p150 ” ) .
  • the ADAR is ADAR2.
  • the ADAR is an ADAR2 expressed by the host cell, e.g., ADAR2 expressed by cerebellum cells.
  • the ADAR is an ADAR exogenous to the host cell. In some embodiments, the ADAR is a hyperactive mutant of a naturally occurring ADAR. In some embodiments, the ADAR is ADAR1 comprising an E1008Q mutation. In some embodiments, the ADAR is not a fusion protein comprising a binding domain. In some embodiments, the ADAR does not comprise an engineered double-strand nucleic acid-binding domain. In some embodiments, the ADAR does not comprise a MCP domain that binds to MS2 hairpin that is fused to the complementary RNA sequence in the dRNA. In some embodiments, the ADAR does not comprise a DSB-binding domain.
  • the host cell has high expression level of ADAR1 (such as ADAR1 p110 and/or ADAR1 p150 ) , e.g., at least about any one of 10%, 20%, 50%, 100%, 2x, 3x, 5x, or more relative to the protein expression level of ⁇ -tubulin.
  • the host cell has high expression level of ADAR2, e.g., at least about any one of 10%, 20%, 50%, 100%, 2x, 3x, 5x, or more relative to the protein expression level of ⁇ -tubulin.
  • the host cell has low expression level of ADAR3, e.g., no more than about any one of 5x, 3x, 2x, 100%, 50%, 20%or less relative to the protein expression level of ⁇ -tubulin.
  • the complementary RNA sequence comprises a cytidine, adenosine or uridine directly opposite the target A in the target RNA.
  • complementary RNA sequence comprises a cytidine mismatch directly opposite the target A in the target RNA.
  • the cytidine mismatch is located at least 5 nucleotides, e.g., at least 10, 15, 20, 25, 30, or more nucleotides, away from the 5’ end of the complementary RNA sequence.
  • the cytidine mismatch is located at least 20 nucleotides, e.g., at least 25, 30, 35, or more nucleotides, away from the 3’ end of the complementary RNA sequence. In some embodiments, the cytidine mismatch is not located within 20 (e.g., 15, 10, 5 or fewer) nucleotides away from the 3’ end of the complementary RNA sequence.
  • the cytidine mismatch is located at least 20 nucleotides (e.g., at least 25, 30, 35, or more nucleotides) away from the 3’ end and at least 5 nucleotides (e.g., at least 10, 15, 20, 25, 30, or more nucleotides) away from the 5’ end of the complementary RNA sequence.
  • the cytidine mismatch is located in the center of the complementary RNA sequence. In some embodiments, the cytidine mismatch is located within 20 nucleotides (e.g., 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotide) of the center of the complementary sequence in the dRNA.
  • the complementary RNA sequence further comprises one or more guanosine (s) , such as 1, 2, 3, 4, 5, 6, or more Gs, that is each directly opposite a non-target adenosine in the target RNA.
  • the complementary RNA sequence comprises two or more consecutive mismatch nucleotides (e.g., 2, 3, 4, 5, or more mismatch nucleotides) opposite a non-target adenosine in the target RNA.
  • the target RNA comprises no more than about 20 non-target As, such as no more than about any one of 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 non-target A.
  • the Gs and consecutive mismatch nucleotides opposite non-target As may reduce off-target editing effects by ADAR.
  • the 5’ nearest neighbor of the target A is a nucleotide selected from U, C, A and G with the preference U>C ⁇ A>G and the 3’ nearest neighbor of the target A is a nucleotide selected from G, C, A and U with the preference G>C>A ⁇ U.
  • the target A is in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA.
  • the three-base motif is UAG
  • the dRNA comprises an A directly opposite the U in the three-base motif, a C directly opposite the target A, and a C, G or U directly opposite the G in the three-base motif.
  • the three-base motif is UAG in the target RNA, and the dRNA comprises ACC, ACG or ACU that is opposite the UAG of the target RNA.
  • the three-base motif is UAG in the target RNA, and the dRNA comprises ACC that is opposite the UAG of the target RNA.
  • the target RNA is any one selected from the group consisting of a pre-messenger RNA, a messenger RNA, a ribosomal RNA, a transfer RNA, a long non-coding RNA and a small RNA (e.g., miRNA) .
  • the target RNA is a pre-messenger RNA.
  • the target RNA is a messenger RNA.
  • the method further comprises introducing an inhibitor of ADAR3 to the host cell.
  • the inhibitor of ADAR3 is an RNAi against ADAR3, such as a shRNA against ADAR3 or a siRNA against ADAR3.
  • the method further comprises introducing a stimulator of interferon to the host cell.
  • the ADAR is inducible by interferon, for example, the ADAR is ADAR p150 .
  • the stimulator of interferon is IFN ⁇ .
  • the inhibitor of ADAR3 and/or the stimulator of interferon are encoded by the same construct (e.g., vector) that encodes the dRNA.
  • the efficiency of editing of the target RNA is at least about 5%, such as at least about any one of 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or higher.
  • the efficiency of editing of the target RNA is at least about 5% (e.g., at least about 7%) in vivo, e.g., in an animal.
  • the efficiency of editing of the target RNA is at least about 10%(e.g., at least about 15%) in a cell in vitro or ex vivo.
  • the efficiency of editing is determined by Sanger sequencing.
  • the efficiency of editing is determined by next-generation sequencing.
  • the method has low off-target editing rate. In some embodiments, the method has lower than about 1% (e.g., no more than about any one of 0.5%, 0.1%, 0.05%, 0.01%, 0.001%or lower) editing efficiency on non-target As in the target RNA. In some embodiments, the method does not edit non-target As in the target RNA. In some embodiments, the method has lower than about 0.1% (e.g., no more than about any one of 0.05%, 0.01%, 0.005%, 0.001%, 0.0001%or lower) editing efficiency on As in non-target RNA.
  • the method does not induce immune response, such as innate immune response. In some embodiments, the method does not induce interferon and/or interleukin expression in the host cell. In some embodiments, the method does not induce IFN- ⁇ and/or IL-6 expression in the host cell.
  • edited RNA or host cells having an edited RNA produced by any one of the methods described herein comprises an inosine.
  • the host cell comprises an RNA having a missense mutation, an early stop codon, an alternative splice site, or an aberrant splice site.
  • the host cell comprises a mutant, truncated, or misfolded protein.
  • the host cell refers to any cell type that can be used as a host cell provided it can be modified as described herein.
  • the host cell may be a host cell with endogenously expressed adenosine deaminase acting on RNA (ADAR) , or may be a host cell into which an adenosine deaminase acting on RNA (ADAR) is introduced by a known method in the art.
  • the host cell may be a prokaryotic cell, a eukaryotic cell or a plant cell.
  • the host cell is derived from a pre-established cell line, such as mammalian cell lines including human cell lines or non-human cell lines.
  • the host cell is derived from an individual, such as a human individual.
  • “Introducing” or “introduction” used herein means delivering one or more polynucleotides, such as dRNAs or one or more constructs including vectors as described herein, one or more transcripts thereof, to a host cell.
  • the invention serves as a basic platform for enabling targeted editing of RNA, for example, pre-messenger RNA, a messenger RNA, a ribosomal RNA, a transfer RNA, a long non-coding RNA and a small RNA (such as miRNA) .
  • the methods of the present application can employ many delivery systems, including but not limited to, viral, liposome, electroporation, microinjection and conjugation, to achieve the introduction of the dRNA or construct as described herein into a host cell.
  • Non-viral vector delivery systems include DNA plasmids, RNA (e.g. a transcript of a construct described herein) , naked nucleic acid, and nucleic acid complexed with a delivery vehicle, such as a liposome.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes for delivery to the host cell.
  • Methods of non-viral delivery of nucleic acids include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid: nucleic acid conjugates, electroporation, nanoparticles, exosomes, microvesicles, or gene-gun, naked DNA and artificial virions.
  • RNA or DNA viral based systems for the delivery of nucleic acids has high efficiency in targeting a virus to specific cells and trafficking the viral payload to the cellular nuclei.
  • the method comprises introducing a viral vector (such as lentiviral vector) encoding the dRNA to the host cell.
  • the viral vector is an AAV, e.g., AAV8.
  • the method comprises introducing a plasmid encoding the dRNA to the host cell.
  • the method comprises introducing (e.g., by electroporation) the dRNA (e.g., synthetic dRNA) into the host cell.
  • the method comprises transfection of the dRNA into the host cell.
  • modification of the target RNA and/or the protein encoded by the target RNA can be determined using different methods depending on the positions of the targeted adenosines in the target RNA. For example, in order to determine whether “A” has been edited to “I” in the target RNA, RNA sequencing methods known in the art can be used to detect the modification of the RNA sequence.
  • the RNA editing may cause changes to the amino acid sequence encoded by the mRNA. For example, point mutations may be introduced to the mRNA of an innate or acquired point mutation in the mRNA may be reversed to yield wild-type gene product (s) because of the conversion of “A” to “I” .
  • Amino acid sequencing by methods known in the art can be used to find any changes of amino acid residues in the encoded protein.
  • Modifications of a stop codon may be determined by assessing the presence of a functional, elongated, truncated, full-length and/or wild-type protein. For example, when the target adenosine is located in a UGA, UAG, or UAA stop codon, modification of the target A (UGA or UAG) or As (UAA) may create a read-through mutation and/or an elongated protein, or a truncated protein encoded by the target RNA may be reversed to create a functional, full-length and/or wild-type protein.
  • Editing of a target RNA may also generate an aberrant splice site, and/or alternative splice site in the target RNA, thus leading to an elongated, truncated, or misfolded protein, or an aberrant splicing or alternative splicing site encoded in the target RNA may be reversed to create a functional, correctly-folding, full-length and/or wild-type protein.
  • the present application contemplates editing of both innate and acquired genetic changes, for example, missense mutation, early stop codon, aberrant splicing or alternative splicing site encoded by a target RNA. Using known methods to assess the function of the protein encoded by the target RNA can find out whether the RNA editing achieves the desired effects.
  • identification of the deamination into inosine may provide assessment on whether a functional protein is present, or whether a disease or drug resistance-associated RNA caused by the presence of a mutated adenosine is reversed or partly reversed.
  • identification of the deamination into inosine may provide a functional indication for identifying a cause of disease or a relevant factor of a disease.
  • the read-out may be the assessment of occurrence and frequency of aberrant splicing.
  • the deamination of a target adenosine is desirable to introduce a splice site, then similar approaches can be used to check whether the required type of splicing occurs.
  • An exemplary suitable method to identify the presence of an inosine after deamination of the target adenosine is RT-PCR and sequencing, using methods that are well-known to the person skilled in the art.
  • target adenosine include, for example, point mutation, early stop codon, aberrant splice site, alternative splice site and misfolding of the resulting protein.
  • These effects may induce structural and functional changes of RNAs and/or proteins associated with diseases, whether they are genetically inherited or caused by acquired genetic mutations, or may induce structural and functional changes of RNAs and/or proteins associated with occurrence of drug resistance.
  • the dRNAs, the constructs encoding the dRNAs, and the RNA editing methods of present application can be used in prevention or treatment of hereditary genetic diseases or conditions, or diseases or conditions associated with acquired genetic mutations by changing the structure and/or function of the disease-associated RNAs and/or proteins.
  • the target RNA is a regulatory RNA.
  • the target RNA to be edited is a ribosomal RNA, a transfer RNA, a long non-coding RNA or a small RNA (e.g., miRNA, pri-miRNA, pre-miRNA, piRNA, siRNA, snoRNA, snRNA, exRNA or scaRNA) .
  • the effects of deamination of the target adenosines include, for example, structural and functional changes of the ribosomal RNA, transfer RNA, long non-coding RNA or small RNA (e.g., miRNA) , including changes of three-dimensional structure and/or loss of function or gain of function of the target RNA.
  • deamination of the target As in the target RNA changes the expression level of one or more downstream molecules (e.g., protein, RNA and/or metabolites) of the target RNA. Changes of the expression level of the downstream molecules can be increase or decrease in the expression level.
  • downstream molecules e.g., protein, RNA and/or metabolites
  • Some embodiments of the present application involve multiplex editing of target RNAs in host cells, which are useful for screening different variants of a target gene or different genes in the host cells.
  • the method comprises introducing a plurality of dRNAs to the host cells, at least two of the dRNAs of the plurality of dRNAs have different sequences and/or have different target RNAs.
  • each dRNA has a different sequence and/or different target RNA.
  • the method generates a plurality (e.g., at least 2, 3, 5, 10, 50, 100, 1000 or more) of modifications in a single target RNA in the host cells.
  • the method generates a modification in a plurality (e.g., at least 2, 3, 5, 10, 50, 100, 1000 or more) of target RNAs in the host cells.
  • the method comprises editing a plurality of target RNAs in a plurality of populations of host cells.
  • each population of host cells receive a different dRNA or a dRNAs having a different target RNA from the other populations of host cells.
  • the present application provides a deaminase-recruiting RNA useful for any one of the methods described herein.
  • Any one of the dRNAs described in this section may be used in the methods of RNA editing and treatment described herein. It is intended that any of the features and parameters described herein for dRNAs can be combined with each other, as if each and every combination is individually described.
  • the dRNAs described herein do not comprise a tracrRNA, crRNA or gRNA used in a CRISPR/Cas system.
  • a deaminase-recruiting RNA for deamination of a target adenosine in a target RNA by recruiting an ADAR, comprising a complementary RNA sequence that hybridizes to the target RNA.
  • the present provides a construct comprising any one of the deaminase-recruiting RNAs described herein.
  • the construct is a viral vector (such as a lentivirus vector) or a plasmid.
  • the viral vector is an AAV, such as AAV8.
  • the construct encodes a single dRNA.
  • the construct encodes a plurality (e.g., about any one of 1, 2, 3, 4, 5, 10, 20 or more) dRNAs.
  • the present application provides a library comprising a plurality of the deaminase-recruiting RNAs or a plurality of the constructs described herein.
  • the present application provides a composition or a host cell comprising the deaminase-recruiting RNA or the construct described herein.
  • the host cell is a prokaryotic cell or a eukaryotic cell.
  • the host cell is a mammalian cell. Most preferably, the host cell is a human cell.
  • the complementary RNA sequence comprises a cytidine, adenosine or uridine directly opposite the target adenosine to be edited in the target RNA.
  • the complementary RNA sequence further comprises one or more guanosine (s) that is each directly opposite a non-target adenosine in the target RNA.
  • the 5’ nearest neighbor of the target A is a nucleotide selected from U, C, A and G with the preference U>C ⁇ A>G and the 3’ nearest neighbor of the target A is a nucleotide selected from G, C, A and U with the preference G>C>A ⁇ U.
  • the 5’ nearest neighbor of the target A is U.
  • the 5’ nearest neighbor of the target A is C or A.
  • the 3’ nearest neighbor of the target A is G.
  • the 3’ nearest neighbor of the target A is C.
  • the target A is in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA.
  • the three-base motif is UAG
  • the dRNA comprises an A directly opposite the U in the three-base motif, a C directly opposite the target A, and a C, G or U directly opposite the G in the three-base motif.
  • the three-base motif is UAG in the target RNA, and the dRNA comprises ACC, ACG or ACU that is opposite the UAG of the target RNA.
  • the dRNA comprises a cytidine mismatch directly opposite the target A in the target RNA.
  • the cytidine mismatch is close to the center of the complementary RNA sequence, such as within 20, 15, 10, 5, 4, 3, 2, or 1 nucleotide away from the center of the complementary RNA sequence.
  • the cytidine mismatch is at least 5 nucleotides away from the 5’ end of the complementary RNA sequence.
  • the cytidine mismatch is at least 20 nucleotides away from the 3’ end of the complementary RNA sequence.
  • the dRNA comprises more than about any one of 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nucleotides.
  • the dRNA is about any one of 40-260, 45-250, 50-240, 60-230, 65-220, 70-210, 70-200, 70-190, 70-180, 70-170, 70-160, 70-150, 70-140, 70-130, 70-120, 70-110, 70-100, 70-90, 70-80, 75-200, 80-190, 85-180, 90-170, 95-160, 100-150 or 105-140 nucleotides in length.
  • the dRNA comprises the nucleic acid sequence of any one of SEQ ID NOs: 25-44, 142-205, 341-342.
  • the dRNA of the present application comprises a complementary RNA sequence that hybridizes to the target RNA.
  • the complementary RNA sequence is perfectly complementary or substantially complementarity to the target RNA to allow hybridization of the complementary RNA sequence to the target RNA.
  • the complementary RNA sequence has 100%sequence complementarity as the target RNA.
  • the complementary RNA sequence is at least about any one of 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%or more complementary to over a continuous stretch of at least about any one of 20, 40, 60, 80, 100, 150, 200, or more nucleotides in the target RNA.
  • the dsRNA formed by hybridization between the complementary RNA sequence and the target RNA has one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) non-Watson-Crick base pairs (i.e., mismatches) .
  • ADAR for example, human ADAR enzymes edit double stranded RNA (dsRNA) structures with varying specificity, depending on a number of factors.
  • dsRNA double stranded RNA
  • One important factor is the degree of complementarity of the two strands making up the dsRNA sequence.
  • Perfect complementarity of between the dRNA and the target RNA usually causes the catalytic domain of ADAR to deaminate adenosines in a non-discriminative manner.
  • the specificity and efficiency of ADAR can be modified by introducing mismatches in the dsRNA region. For example, A-C mismatch is preferably recommended to increase the specificity and efficiency of deamination of the adenosine to be edited.
  • the G-A mismatch can reduce off-target editing.
  • Perfect complementarity is not necessarily required for a dsRNA formation between the dRNA and its target RNA, provided there is substantial complementarity for hybridization and formation of the dsRNA between the dRNA and the target RNA.
  • the dRNA sequence or single-stranded RNA region thereof has at least about any one of 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%of sequence complementarity to the target RNA, when optimally aligned.
  • Optimal alignment may be determined with the use of any suitable algorithm for aligning sequences, non-limiting examples of which include the Smith-Waterman algorithm, the Needleman-Wimsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g. the Burrows Wheeler Aligner) .
  • any suitable algorithm for aligning sequences non-limiting examples of which include the Smith-Waterman algorithm, the Needleman-Wimsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g. the Burrows Wheeler Aligner) .
  • the nucleotides neighboring the target adenosine also affect the specificity and efficiency of deamination.
  • the 5’ nearest neighbor of the target adenosine to be edited in the target RNA sequence has the preference U>C ⁇ A>G and the 3’ nearest neighbor of the target adenosine to be edited in the target RNA sequence has the preference G>C>A ⁇ U in terms of specificity and efficiency of deamination of adenosine.
  • the target adenosine when the target adenosine may be in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA, the specificity and efficiency of deamination of adenosine are higher than adenosines in other three-base motifs.
  • the target adenosine to be edited is in the three-base motifUAG, UAC, UAA, UAU, CAG, CAC, AAG, AAC or AAA
  • the efficiency of deamination of adenosine is much higher than adenosines in other motifs.
  • different designs of dRNA may also lead to different deamination efficiency.
  • the dRNA comprises cytidine (C) directly opposite the target adenosine to be edited, adenosine (A) directly opposite the uridine, and cytidine (C) , guanosine (G) or uridine (U) directly opposite the guanosine
  • the efficiency of deamination of the target adenosine is higher than that using other dRNA sequences.
  • the editing efficiency of the A in the UAG of the target RNA may reach about 25%-30%.
  • dRNAs can be designed to comprise one or more guanosines directly opposite one or more adenosine (s) other than the target adenosine to be edited in the target RNA.
  • mismatch refers to opposing nucleotides in a double stranded RNA (dsRNA) which do not form perfect base pairs according to the Watson-Crick base pairing rules. Mismatch base pairs include, for example, G-A, C-A, U-C, A-A, G-G, C-C, U-U base pairs.
  • a dRNA is designed to comprise a C opposite the A to be edited, generating a A-C mismatch in the dsRNA formed by hybridization between the target RNA and dRNA.
  • the dsRNA formed by hybridization between the dRNA and the target RNA does not comprise a mismatch.
  • the dsRNA formed by hybridization between the dRNA and the target RNA comprises one or more, such as any one of 1, 2, 3, 4, 5, 6, 7 or more mismatches (e.g., the same type of different types of mismatches) .
  • the dsRNA formed by hybridization between the dRNA and the target RNA comprises one or more kinds of mismatches, for example, 1, 2, 3, 4, 5, 6, 7 kinds of mismatches selected from the group consisting of G-A, C-A, U-C, A-A, G-G, C-C and U-U.
  • the mismatch nucleotides in the dsRNA formed by hybridization between the dRNA and the target RNA can form bulges, which can promote the efficiency of editing of the target RNA.
  • the additional bulge-inducing mismatches may be upstream and/or downstream of the target adenosine.
  • the bulges may be single-mismatch bulges (caused by one mismatching base pair) or multi-mismatch bulges (caused by more than one consecutive mismatching base pairs, preferably two or three consecutive mismatching base pairs) .
  • the complementary RNA sequence in the dRNA is single-stranded.
  • the dRNA may be entirely single-stranded or have one or more (e.g., 1, 2, 3, or more) double-stranded regions and/or one or more stem loop regions.
  • the complementary RNA sequence is at least about any one of 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200 or more nucleotides.
  • the complementary RNA sequence is about any one of 40-260, 45-250, 50-240, 60-230, 65-220, 70-220, 70-210, 70-200, 70-190, 70-180, 70-170, 70-160, 70-150, 70-140, 70-130, 70-120, 70-110, 70-100, 70-90, 70-80, 75-200, 80-190, 85-180, 90-170, 95-160, 100-200, 100-150, 100-175, 110-200, 110-175, 110-150, or 105-140 nucleotides in length.
  • the complementary RNA sequence is about 71 nucleotides long. In some embodiments, the complementary RNA sequence is about 111 nucleotides long.
  • the dRNA apart from the complementary RNA sequence, further comprises regions for stabilizing the dRNA, for example, one or more double-stranded regions and/or stem loop regions.
  • the double-stranded region or stem loop region of the dRNA comprises no more than about any one of 200, 150, 100, 50, 40, 30, 20, 10 or fewer base-pairs.
  • the dRNA does not comprise a stem loop or double-stranded region.
  • the dRNA comprises an ADAR-recruiting domain. In some embodiments, the dRNA does not comprise an ADAR-recruiting domain.
  • the dRNA may comprise one or more modifications.
  • the dRNA has one or more modified nucleotides, including nucleobase modification and/or backbone modification.
  • modified nucleotides including nucleobase modification and/or backbone modification.
  • Exemplary modifications to the RNA include, but are not limited to, phosphorothioate backbone modification, 2’ -substitutions in the ribose (such as 2’ -O-methyl and 2’ -fluoro substitutions) , LNA, and L-RNA.
  • the dRNA does not have modifications to the nucleobase or backbone.
  • construct refers to DNA or RNA molecules that comprise a coding nucleotide sequence that can be transcribed into RNAs or expressed into proteins.
  • the construct contains one or more regulatory elements operably linked to the nucleotide sequence encoding the RNA or protein.
  • the construct comprises a promoter that is operably linked, or spatially connected to the coding nucleotide sequence, such that the promoter controls the transcription or expression of the coding nucleotide sequence.
  • a promoter may be positioned 5' (upstream) of a coding nucleotide sequence under its control.
  • the distance between the promoter and the coding sequence may be approximately the same as the distance between that promoter and the gene it controls in the gene from which the promoter is derived. As is known in the art, variation in this distance may be accommodated without loss of promoter function.
  • the construct comprises a 5’ UTR and/or a 3’ UTR that regulates the transcription or expression of the coding nucleotide sequence.
  • the construct is a vector encoding any one of the dRNAs disclosed in the present application.
  • the term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • Vectors include, but are not limited to, nucleic acid molecules that are single-stranded, double-stranded, or partially double-stranded; nucleic acid molecules that comprise one or more free ends, no free ends (e.g. circular) ; nucleic acid molecules that comprise DNA, RNA, or both; and other varieties of polynucleotides known in the art.
  • vector refers to a circular double stranded DNA loop into which additional DNA segments can be inserted, such as by standard molecular cloning techniques.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors) .
  • Other vectors e.g., non-episomal mammalian vectors
  • certain vectors are capable of directing the transcription or expression of coding nucleotide sequences to which they are operatively linked. Such vectors are referred to herein as “expression vectors” .
  • Recombinant expression vectors can comprise a nucleic acid of the invention in a form suitable for transcription or expression of the nucleic acid in a host cell.
  • the recombinant expression vector includes one or more regulatory elements, which may be selected on the basis of the host cells to be used for transcription or expression, which is operatively linked to the nucleic acid sequence to be transcribed or expressed.
  • “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory element (s) in a manner that allows for expression of the nucleotide sequence (e.g. in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell) .
  • a construct comprising a nucleotide sequence encoding the dRNA.
  • a construct comprising a nucleotide sequence encoding the ADAR.
  • a construct comprising a first nucleotide sequence encoding the dRNA and a second nucleotide sequence encoding the ADAR.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to the same promoter.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to different promoters.
  • the promoter is inducible.
  • the construct does not encode for the ADAR.
  • the vector further comprises nucleic acid sequence (s) encoding an inhibitor of ADAR3 (e.g., ADAR3 shRNA or siRNA) and/or a stimulator of interferon (e.g., IFN- ⁇ ) .
  • the construct is an AAV, such as AAV8.
  • RNA editing methods and compositions described herein may be used to treat or prevent a disease or condition in an individual, including, but not limited to hereditary genetic diseases and drug resistance.
  • a method of editing a target RNA in a cell of an individual comprising editing the target RNA using any one of the methods of RNA editing described herein.
  • a method of editing a target RNA in a cell of an individual comprising introducing a dRNA or a construct encoding the dRNA into the cell of the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA.
  • the target RNA is associated with a disease or condition of the individual.
  • the disease or condition is a hereditary genetic disease or a disease or condition associated with one or more acquired genetic mutations (e.g., drug resistance) .
  • the method further comprises obtaining the cell from the individual.
  • a method of restoring expression or activity of a target RNA or protein encoded by the target RNA in a cell of an individual having a disease or condition comprising editing the target RNA using any one of the methods of RNA editing described herein, wherein the target RNA has a G to A mutation associated with the disease or condition.
  • the method is carried out on an isolated cell from an individual ex vivo. In some embodiments, the method is carried out in vivo.
  • a method of treating or preventing a disease or condition in an individual comprising editing a target RNA associated with the disease or condition in a cell of the individual using any one of the methods of RNA editing described herein.
  • a method of treating or preventing a disease or condition in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to a target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the method further comprises culturing the cell having the edited RNA. In some embodiments, the method further comprises administering the cell having the edited RNA to the individual.
  • the disease or condition is a hereditary genetic disease or a disease or condition associated with one or more acquired genetic mutations (e.g., drug resistance) .
  • a method of treating or preventing a disease or condition in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to a target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an endogenously expressed ADAR of the host cell to deaminate a target A in the target RNA.
  • the method further comprises culturing the cell having the edited RNA.
  • the method further comprises administering the cell having the edited RNA to the individual.
  • the disease or condition is a hereditary genetic disease or a disease or condition associated with one or more acquired genetic mutations (e.g., drug resistance) .
  • a method of treating or preventing a disease or condition in an individual comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to a target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the disease or condition is a hereditary genetic disease or a disease or condition associated with one or more acquired genetic mutations (e.g., drug resistance) .
  • Diseases and conditions suitable for treatment using the methods of the present application include diseases associated with a mutation, such as a G to A mutation, e.g., a G to A mutation that results in missense mutation, early stop codon, aberrant splicing, or alternative splicing in an RNA transcript.
  • a mutation such as a G to A mutation, e.g., a G to A mutation that results in missense mutation, early stop codon, aberrant splicing, or alternative splicing in an RNA transcript.
  • TP53 W53X e.g., 158G>A
  • IDUA W402X e.g., TGG>TAG mutation in exon 9
  • Mucopolysaccharidosis type I MPS I
  • COL3A1 W1278X e.g., 3833G>A mutation
  • BMPR2 W298X e.g., 893G>A
  • AHI1 W725X e.g., 2174G>A
  • FANCC W506X e.g., 1517G>A
  • MYBPC3 W1098X e.g., 3293G>A
  • primary familial hypertrophic cardiomyopathy e.g., 7
  • a method of treating a cancer associated with a target RNA having a mutation comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is TP53 W53X (e.g., 158G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 195, 196 or 197.
  • a method of treating or preventing a cancer with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is TP53 W53X (e.g., 158G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 195, 196 or 197.
  • a method of treating MPS I e.g., Hurler syndrome or Scheie syndrome
  • a target RNA having a mutation e.g., G>A mutation
  • the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is IDUA W402X (e.g., TGG>TAG mutation in exon 9) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 204 or 205.
  • a method of treating or preventing MPS I with a target RNA having a mutation (e.g., G>A mutation) in an individual, comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is IDUA W402X (e.g., TGG>TAG mutation in exon 9) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 204 or 205.
  • a method of treating or preventing Ehlers-Danlos syndrome associated with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is COL3A1 W1278X (e.g., 3833G>A mutation) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 198.
  • a method of treating or preventing Ehlers-Danlos syndrome with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is COL3A1 W1278X (e.g., 3833G>A mutation) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 198.
  • a method of treating primary pulmonary hypertension associated with a target RNA having a mutation comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is BMPR2 W298X (e.g., 893G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 199.
  • a method of treating or preventing primary pulmonary hypertension with a target RNA having a mutation comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is BMPR2 W298X (e.g., 893G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 199.
  • a method of treating Joubert syndrome associated with a target RNA having a mutation comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is AHI1 W725X (e.g., 2174G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 200.
  • a method of treating or preventing Joubert syndrome with a target RNA having a mutation comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is AHI1 W725X (e.g., 2174G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 200.
  • a method of treating Fanconi anemia associated with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is FANCC W506X (e.g., 1517G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 201.
  • a method of treating or preventing Fanconi anemia with a target RNA having a mutation comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is FANCC W506X (e.g., 1517G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 201.
  • a method of treating primary familial hypertrophic cardiomyopathy associated with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is MYBPC3 W1098X (e.g., 3293G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 202.
  • a method of treating or preventing primary familial hypertrophic cardiomyopathy with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is MYBPC3 W1098X (e.g., 3293G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 202.
  • a method of treating X-linked severe combined immunodeficiency associated with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising introducing a dRNA or a construct encoding the dRNA into an isolated cell of the individual ex vivo, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the isolated cell.
  • the method comprises introducing the ADAR or a construct encoding the ADAR to the isolated cell.
  • the target RNA is IL2RG W237X (e.g., 710G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 203.
  • a method of treating or preventing X-linked severe combined immunodeficiency with a target RNA having a mutation (e.g., G>A mutation) in an individual comprising administering an effective amount of a dRNA or a construct encoding the dRNA to the individual, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA associated with the disease or condition, and wherein the dRNA is capable of recruiting an ADAR to deaminate a target A in the target RNA, thereby rescuing the mutation in the target RNA.
  • the ADAR is an endogenously expressed ADAR in the cells of the individual.
  • the method comprises administering the ADAR or a construct encoding the ADAR to the individual.
  • the target RNA is IL2RG W237X (e.g., 710G>A) .
  • the dRNA comprises the nucleic acid sequence of SEQ ID NO: 203.
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: decreasing one more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease) , preventing or delaying the spread (e.g., metastasis) of the disease, preventing or delaying the occurrence or recurrence of the disease, delay or slowing the progression of the disease, ameliorating the disease state, providing a remission (whether partial or total) of the disease, decreasing the dose of one or more other medications required to treat the disease, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival.
  • treatment is a reduction of pathological consequence of the disease or condition. The methods of the invention contemplate any one or more of these aspects of treatment.
  • the terms “individual, ” “subject” and “patient” are used interchangeably herein to describe a mammal, including humans.
  • An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate.
  • the individual is human.
  • an individual suffers from a disease or condition, such as drug resistance.
  • the individual is in need of treatment.
  • an “effective amount” refers to an amount of a composition (e.g., dRNA or constructs encoding the dRNA) sufficient to produce a desired therapeutic outcome (e.g., reducing the severity or duration of, stabilizing the severity of, or eliminating one or more symptoms of a disease or condition) .
  • beneficial or desired results include, e.g., decreasing one or more symptoms resulting from the disease (biochemical, histologic and/or behavioral) , including its complications and intermediate pathological phenotypes presented during development of the disease, increasing the quality of life of those suffering from the disease or condition, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, and/or prolonging survival of patients.
  • the disease biochemical, histologic and/or behavioral
  • beneficial or desired results include, e.g., decreasing one or more symptoms resulting from the disease (biochemical, histologic and/or behavioral) , including its complications and intermediate pathological phenotypes presented during development of the disease, increasing the quality of life of those suffering from the disease or condition, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, and/or prolonging survival of patients.
  • dosages, schedules, and routes of administration of the compositions may be determined according to the size and condition of the individual, and according to standard pharmaceutical practice.
  • routes of administration include intravenous, intra-arterial, intraperitoneal, intrapulmonary, intravesicular, intramuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, or transdermal.
  • RNA editing methods of the present application can not only be used in animal cells, for example mammalian cells, but also may be used in modification of RNAs of plant or fungi, for example, in plants or fungi that have endogenously expressed ADARs.
  • the methods described herein can be used to generate genetically engineered plant and fungi with improved properties.
  • compositions are compositions, kits and articles of manufacture
  • compositions comprising any one of the dRNAs, constructs, libraries, or host cells having edited RNA as described herein.
  • a pharmaceutical composition comprising any one of the dRNAs or constructs encoding the dRNA described herein, and a pharmaceutically acceptable carrier, excipient or stabilizer.
  • a pharmaceutically acceptable carrier, excipients and stabilizers have been described, for example, in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980) .
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propylparaben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol) ; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as olyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • compositions to be used for in vivo administration must be sterile. This is readily accomplished by, e.g., filtration through sterile filtration membranes. Further provided are kits or articles of manufacture useful for any one of the methods of RNA editing or methods of treatment described herein, comprising any one of the dRNAs, constructs, compositions, libraries, or edited host cells as described herein.
  • kits for editing a target RNA in a host cell comprising a dRNA, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, wherein the dRNA is capable of recruiting an ADAR to deaminate an A in the target RNA.
  • the kit further comprises an ADAR or a construct encoding an ADAR.
  • the kit further comprises an inhibitor of ADAR3 or a construct thereof.
  • the kit further comprises a stimulator of interferon or a construct thereof.
  • the kit further comprises an instruction for carrying out any one of the RNA editing methods described herein.
  • kits of the present application are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags) , and the like. Kits may optionally provide additional components such as transfection or transduction reagents, cell culturing medium, buffers, and interpretative information.
  • the present application thus also provides articles of manufacture.
  • the article of manufacture can comprise a container and a label or package insert on or associated with the container.
  • Suitable containers include vials (such as sealed vials) , bottles, jars, flexible packaging, and the like.
  • the container holds a pharmaceutical composition, and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle) .
  • the container holding the pharmaceutical composition may be a multi-use vial, which allows for repeat administrations (e.g. from 2-6 administrations) of the reconstituted formulation.
  • Package insert refers to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such products.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI) , phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • kits or article of manufacture may include multiple unit doses of the pharmaceutical compositions and instructions for use, packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies.
  • a method for editing a target RNA in a host cell comprising introducing a deaminase-recruiting RNA (dRNA) or a construct encoding the dRNA into the host cell, wherein the dRNA comprises a complementary RNA sequence that hybridizes to the target RNA, and wherein the deaminase-recruiting RNA is capable of recruiting an adenosine deaminase acting on RNA (ADAR) to deaminate a target adenosine in the target RNA.
  • dRNA deaminase-recruiting RNA
  • ADAR adenosine deaminase acting on RNA
  • RNA sequence comprises a cytidine, adenosine or uridine directly opposite the target adenosine in the target RNA.
  • RNA sequence comprises a cytidine mismatch directly opposite the target adenosine in the target RNA.
  • RNA sequence further comprises one or more guanosines each opposite a non-target adenosine in the target RNA.
  • the target adenosine is in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA.
  • the deaminase-recruiting RNA comprises an A directly opposite the uridine in the three-base motif, a cytidine directly opposite the target adenosine, and a cytidine, guanosine or uridine directly opposite the guanosine in the three-base motif.
  • dRNA is about 100 to about 150 (e.g., about 110-150) nucleotides in length.
  • the target RNA is an RNA selected from the group consisting of a pre-messenger RNA, a messenger RNA, a ribosomal RNA, a transfer RNA, a long non-coding RNA and a small RNA.
  • dRNA does not comprise an ADAR-recruiting domain (e.g., a DSB-binding domain, a GluR2 domain, or a MS2 domain) .
  • ADAR-recruiting domain e.g., a DSB-binding domain, a GluR2 domain, or a MS2 domain
  • dRNA does not comprise a chemically modified nucleotide (e.g., 2’ -O-methyl or phosphorothioate modification) .
  • a chemically modified nucleotide e.g., 2’ -O-methyl or phosphorothioate modification
  • a method for treating or preventing a disease or condition in an individual comprising editing a target RNA associated with the disease or condition in a cell of the individual according to any one of the embodiments 1-39.
  • the target RNA is TP53, and the disease or condition is cancer
  • the target RNA is IDUA, and the disease or condition is Mucopolysaccharidosis type I (MPS I) ;
  • the target RNA is COL3A1, and the disease or condition is Ehlers-Danlos syndrome;
  • the target RNA is BMPR2, and the disease or condition is Joubert syndrome;
  • the target RNA is FANCC, and the disease or condition is Fanconi anemia;
  • the target RNA is MYBPC3, and the disease or condition is primary familial hypertrophic cardiomyopathy; or
  • the target RNA is IL2RG, and the disease or condition is X-linked severe combined immunodeficiency.
  • dRNA deaminase-recruiting RNA
  • RNA sequence comprises a cytosine, adenosine or U directly opposite the target adenosine in the target RNA.
  • RNA sequence comprises a cytidine mismatch directly opposite the target adenosine in the target RNA.
  • dRNA of embodiment 49 wherein the cytidine mismatch is located at least 20 nucleotides away from the 3’ end of the complementary sequence, and at least 5 nucleotides away from the 5’ end of the complementary sequence in the dRNA.
  • dRNA of embodiment 50 wherein the cytidine mismatch is located within 10 nucleotides from the center (e.g., at the center) of the complementary sequence in the dRNA.
  • RNA sequence further comprises one or more guanosines each directly opposite a non-target adenosine in the target RNA.
  • RNA deaminase-recruiting RNA of any one of embodiments 47-53, wherein the target adenosine is in a three-base motif selected from the group consisting of UAG, UAC, UAA, UAU, CAG, CAC, CAA, CAU, AAG, AAC, AAA, AAU, GAG, GAC, GAA and GAU in the target RNA.
  • RNA of embodiment 54 wherein the three-base motif is UAG, and wherein the dRNA comprises an adenosine directly opposite the uridine in the three-base motif, a cytosine directly opposite the target adenosine, and a cytidine, guanosine or uridine directly opposite the guanosine in the three-base motif.
  • deaminase-recruiting RNA of embodiment 55 wherein the three-base motif is UAG in the target RNA, and wherein the deaminase-recruiting RNA comprises ACC, ACG or ACU opposite the UAG of the target RNA.
  • dRNA of embodiment 57 wherein the dRNA is more than about 70 nucleotides in length.
  • dRNA of embodiment 57 or 58, wherein the dRNA is about 100 to about 150 nucleotides (e.g., about 110-150) in length.
  • an ADAR-recruiting domain e.g., a DSB-binding domain, a GluR2 domain, or a MS2 domain
  • a chemically modified nucleotide e.g., 2’ -O-methyl or phosphorothioate modification
  • construct of embodiment 62, wherein the construct is a viral vector (e.g., lentiviral vector) or a plasmid.
  • a library comprising a plurality of the deaminase-recruiting RNAs of any one of embodiments 47-61 or the construct of embodiment 62 or 63.
  • composition comprising the deaminase-recruiting RNA of any one of embodiments 47-61, the construct of embodiment 62 or 63, or the library of embodiment 64.
  • a host cell comprising the deaminase-recruiting RNA of any one of embodiments 47-61 or the construct of embodiment 62 or 63.
  • a kit for editing a target RNA in a host cell comprising a deaminase-recruiting RNA, wherein the deaminase-recruiting RNA comprises a complementary RNA sequence that hybridizes to the target RNA, wherein the deaminase-recruiting RNA is capable of recruiting an ADAR to deaminate a target adenosine in the target RNA.
  • the dual fluorescence reporter was cloned by PCR amplifying mCherry and EGFP (the EGFP first codon ATG was deleted) coding DNA, the 3 ⁇ GS linker and targeting DNA sequence were added via primers during PCR. Then the PCR products were cleaved and linked by Type IIs restriction enzyme BsmB1 (Thermo) and T4 DNA ligase (NEB) , which then were inserted into pLenti backbone (pLenti-CMV-MCS-SV-Bsd, Stanley Cohen Lab, Stanford University) .
  • BsmB1 Thermo
  • NEB T4 DNA ligase
  • the dLbuCas13 DNA was PCR amplified from the Lbu plasmids (Addgene #83485) .
  • the ADAR1DD and ADAR2DD were amplified from Adar1 (p150) cDNA and Adar2 cDNA, both of which were gifts from Han’s lab at Xiamen University.
  • the ADAR1DD or ADAR2DD were fused to dLbuCas13 DNA by overlap-PCR, and the fused PCR products were inserted into pLenti backbone.
  • dRNA sequences were directly synthesized (for short dRNAs) and annealed or PCR amplified by synthesizing overlapping ssDNA, and the products were cloned into the corresponding vectors under U6 expression by Golden-gate cloning.
  • Adar1 and Adar1 were PCR amplified from Adar1 (p150) cDNA, and the full length Adar2 were PCR amplified from Adar2 cDNA, which were then cloned into pLenti backbone, respectively.
  • mCherry and EGFP (the start codon ATG of EGFP was deleted) coding sequences were PCR amplified, digested using BsmBI (Thermo Fisher Scientific, ER0452) , followed by T4 DNA ligase (NEB, M0202L) -mediated ligation with GGGGS linkers. The ligation product was subsequently inserted into the pLenti-CMV-MCS-PURO backbone.
  • the ADAR1 DD gene was amplified from the ADAR1 p150 construct (a gift from Jiahuai Han’s lab, Xiamen University) .
  • the dLbuCas13 gene was amplified by PCR from the Lbu_C2c2_R472A_H477A_R1048A_H1053A plasmid (Addgene#83485) .
  • the ADAR1 DD hyperactive E1008Q variant was generated by overlap-PCR and then fused to dLbuCas13.
  • the ligation products were inserted into the pLenti-CMV-MCS-BSD backbone.
  • arRNA-expressing construct the sequences of arRNAs were synthesized and golden-gate cloned into the pLenti-sgRNA-lib 2.0 (Addgene#89638) backbone, and the transcription of arRNA was driven by hU6 promoter.
  • the full length ADAR1 p110 and ADAR1 p150 were PCR amplified from the ADAR1 p150 construct
  • the full length ADAR2 were PCR amplified from the ADAR2 construct (a gift from Jiahuai Han’s lab, Xiamen University) .
  • the amplified products were then cloned into the pLenti-CMV-MCS-BSD backbone.
  • TP53 ordered from Vigenebio
  • other 6 disease-relevant genes (COL3A1, BMPR2, AHI1, FANCC, MYBPC3 and IL2RG, gifts from Jianwei Wang’s lab, Institute of pathogen biology, Chinese Academy of Medical Sciences) were amplified from the constructs encoding the corresponding genes with introduction of G>A mutations through mutagenesis PCR.
  • the amplified products were cloned into the pLenti-CMV-MCS-mCherry backbone through Gibson cloning method 59 .
  • Mammalian cell lines were cultured Dulbecco’s Modified Eagle Medium (10-013-CV, Corning, Tewksbury, MA, USA) , adding 10%fetal bovine serum (Lanzhou Bailing Biotechnology Co., Ltd., Lanzhou, China) , supplemented with 1%penicillin–streptomycin under 5%CO 2 at 37°C.
  • the Adar1-KO cell line was purchased from EdiGene China, and the genotyping results were also provided by EdiGene China.
  • the HeLa and B16 cell lines were from Z. Jiang’s laboratory (Peking University) . And the HEK293T cell line was from C. Zhang’s laboratory (Peking University) .
  • RD cell line was from J Wang's laboratory (Institute of Pathogen Biology, Peking Union Medical College&Chinese Academy of Medical Sciences) .
  • SF268 cell lines were from Cell Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences.
  • A549 and SW13 cell lines were from EdiGene Inc. HepG2, HT29, NIH3T3, and MEF cell lines were maintained in our laboratory at Peking University.
  • the human primary pulmonary fibroblasts (#3300) and human primary bronchial epithelial cells (#3210) were purchased from ScienCell Research Laboratories, Inc. and were cultured in Fibroblast Medium (ScienCell, #2301) and Bronchial Epithelial Cell Medium (ScienCell, #3211) , respectively. Both media were supplemented with 15%fetal bovine serum (BI) and 1%penicillin–streptomycin.
  • the primary GM06214 and GM01323 cells were ordered from Coriell Institute for Medical Research and cultured in Dulbecco’s Modified Eagle Medium (Corning, 10-013-CV) with 15%fetal bovine serum (BI) and 1%penicillin–streptomycin. All cells were cultured under 5%CO 2 at 37°C.
  • 293T-WT cells or 293T-Adar1-KO cells were seeded in 6 wells plates (6 ⁇ 10 5 cells/well) , 24 hours later, 1.5 ⁇ g reporter plasmids and 1.5 ⁇ g dRNA plasmids were co-transfected using the X-tremeGENE HP DNA transfection reagent (06366546001; Roche, Mannheim, German) , according to the supplier’s protocols. 48 to 72 hours later, collected cells and performed FACS analysis.
  • Adar1 p110
  • Adar1 p150
  • Adar2 rescue and overexpression experiments 293T-WT cells or 293T-Adar1-KO cells were seeded in 12 wells plates (2.5 ⁇ 10 5 cells/well) , 24 hours later, 0.5 ⁇ g reporter plasmids, 0.5 ⁇ g dRNA plasmids and 0.5 ⁇ g Adar1/2 plasmids (pLenti backbone as control) were co-transfected using the X-tremeGENE HP DNA transfection reagent (06366546001, Roche, Mannheim, German) . 48 to 72 hours later, collected cells and performed FACS analysis.
  • RNA isolation For endogenous mRNA experiments, 293T-WT cells were seeded in 6 wells plates (6 ⁇ 10 5 cells/well) , When approximately 70%confluent, 3 ⁇ g dRNA plasmids were transfected using the X-tremeGENE HP DNA transfection reagent (06366546001, Roche, Mannheim, German) . 72 hours later, collected cells and sorted GFP-positive or BFP-positive cells (according to the corresponding fluorescence maker) via FACS for the following RNA isolation.
  • X-tremeGENE HP DNA transfection reagent 6366546001, Roche, Mannheim, German
  • PBMCs Peripheral blood mononuclear cells
  • T cells were isolated by Ficoll centrifugation (Dakewei, AS1114546)
  • T cells were isolated by magnetic negative selection using an EasySep Human T Cell Isolation Kit (STEMCELL, 17951) from PBMCs.
  • T cells were cultured in X-vivo15 medium, 10%FBS and IL2 (1000 U/ml) and stimulated with CD3/CD28 DynaBeads (ThermoFisher, 11131D) for 2 days.
  • Leukapheresis products from healthy donors were acquired from AllCells LLC China. All healthy donors provided informed consent.
  • the expression plasmid was co-transfected into HEK293T-WT cells, together with two viral packaging plasmids, pR8.74 and pVSVG (Addgene) via the X-tremeGENE HP DNA transfection reagent. 72 hours later, the supernatant virus was collected and stored at-80°C.
  • the HEK293T-WT cells were infected with lenti-virus, 72 hours later, mCherry-positive cells were sorted via FACS and cultured to select a single clone cell lines stably expressing dual fluorescence reporter system with much low EGFP background by limiting dilution method.
  • the reporter constructs (pLenti-CMV-MCS-PURO backbone) were co-transfected into HEK293T cells, together with two viral packaging plasmids, pR8.74 and pVSVG. 72 hours later, the supernatant virus was collected and stored at -80°C.
  • the HEK293T cells were infected with lentivirus, then mCherry-positive cells were sorted via FACS and cultured to select a single clone cell lines stably expressing dual fluorescence reporter system without detectable EGFP background.
  • the HEK293T ADAR1 –/– and TP53 –/– cell lines were generated according to a previously reported method 60 .
  • ADAR1-targeting sgRNA and PCR amplified donor DNA containing CMV-driven puromycin resistant gene were co-transfected into HEK293T cells. Then cells were treated with puromycin 7 days after transfection. Single clones were isolated from puromycin resistant cells followed by verification through sequencing and Western blot.
  • HEK293T cells or HEK293T ADAR1 –/– cells were seeded in 6-well plates (6 ⁇ 10 5 cells/well) . 24 hours later, cells were co-transfected with 1.5 ⁇ g reporter plasmids and 1.5 ⁇ g arRNA plasmids.
  • ADAR1 p110 , ADAR1 p150 or ADAR2 protein expression the editing efficiency was assayed by EGFP positive ratio and deep sequencing.
  • HEK293T ADAR1 –/– cells were seeded in 12-well plates (2.5 ⁇ 10 5 cells/well) . 24 hours later, cells were co-transfected with 0.5 ⁇ g of reporter plasmids, 0.5 ⁇ g arRNA plasmids and 0.5 ⁇ g ADAR1/2 plasmids (pLenti backbone as control) . The editing efficiency was assayed by EGFP positive ratio and deep sequencing.
  • HEK293T cells were seeded in 6-well plates (6 ⁇ 10 5 cells/well) . Twenty-four hours later, cells were transfected with 3 ⁇ g of arRNA plasmids. The editing efficiency was assayed by deep sequencing.
  • RNA editing efficiency in multiple cell lines 8-9 ⁇ 104 (RD, SF268, HeLa) or 1.5 ⁇ 10 5 (HEK293T) cells were seeded in 12-well plates.
  • RD RD
  • SF268, HeLa 1.5 ⁇ 10 5
  • 2-2.5 ⁇ 10 5 cells were seeded in 6-well plate. Twenty-four hours later, reporters and arRNAs plasmid were co-transfected into these cells. The editing efficiency was assayed by EGFP positive ratio.
  • EGFP positive ratio At 48 to 72 hrs post transfection, cells were sorted and collected by Fluorescence-activated cell sorting (FACS) analysis.
  • FACS Fluorescence-activated cell sorting
  • the mCherry signal was served as a fluorescent selection marker for the reporter/arRNA-expressing cells, and the percentages of EGFP + /mCherry + cells were calculated as the readout for editing efficiency.
  • RNA isolation TIANGEN, DP420
  • the total RNAs were reverse-transcribed into cDNA via RT-PCR (TIANGEN, KR103-04) , and the targeted locus was PCR amplified with the corresponding primers listed in the following table.
  • the PCR products were purified for Sanger sequencing or NGS (Illumina HiSeq X Ten) .
  • HEK293 cells were seeded on 6 wells plates (6 ⁇ 10 5 cells/well) .
  • HEK293 cells were transfected with 3 ⁇ g dRNA using the X-tremeGENE HP DNA transfection reagent (Roche) .
  • sorted GFP-positive or BFP-positive cells (according to the corresponding fluorescence marker) via FACS, followed by RNA isolation.
  • the isolated RNA was reverse-transcribed into cDNA via RT-PCR, and specific targeted gene locus were amplified with the corresponding primer pairs (23 PCR cycles) and sequenced on an Illumina NextSeq.
  • HEK293T positive control
  • HEK293T ADAR1 -/- negative control
  • NIH3T3 seven human cell lines
  • RD, HeLa, SF268, A549, HepG2, HT-29, SW13 human cell lines
  • HEK293T 1.5 x 10 5
  • an index was generated using the targeted site sequence (upstream and downstream 20-nt) of arRNA covering sequences. Reads were aligned and quantified using BWA version 0.7.10-r789. Alignment BAMs were then sorted by Samtools, and RNA editing sites were analyzed using REDitools version 1.0.4. The parameters are as follows: -U [AG or TC]-t 8-n 0.0-T 6-6-e-d-u. All the significant A>G conversion within arRNA targeting region calculated by Fisher’s exact test (p-value ⁇ 0.05) were considered as edits by arRNA. The conversions except for targeted adenosine were off-target edits. The mutations that appeared in control and experimental groups simultaneously were considered as SNP.
  • the Ctrl RNA 151 or arRNA 151 -PPIB-expressing plasmids with BFP expression cassette were transfected into HEK293T cells.
  • the BFP + cells were enriched by FACS 48 hours after transfection, and RNAs were purified with RNAprep Pure Micro kit (TIANGEN, DP420) .
  • the mRNA was then purified using NEBNext Poly (A) mRNA Magnetic Isolation Module (New England Biolabs, E7490) , processed with the NEBNext Ultra II RNA Library Prep Kit for Illumina (New England Biolabs, E7770) , followed by deep sequencing analysis using Illumina HiSeq X Ten platform (2 ⁇ 150-bp paired end; 30G for each sample) .
  • NEBNext Poly (A) mRNA Magnetic Isolation Module New England Biolabs, E7490
  • NEBNext Ultra II RNA Library Prep Kit for Illumina New England Biolabs, E7770
  • Ten platform (2 ⁇ 150-bp
  • the bioinformatics analysis pipeline was referred to the work by Vogel et al 22 .
  • the quality control of analysis was conducted by using FastQC, and quality trim was based on Cutadapt (the first 6-bp for each reads were trimmed and up to 20-bp were quality trimmed) .
  • AWK scripts were used to filtered out the introduced arRNAs. After trimming, reads with lengths less than 90-nt were filtered out. Subsequently, the filtered reads were mapped to the reference genome (GRCh38-hg38) by STAR software 61 .
  • the raw VCF files generated by GATK were filtered and annotated by GATK VariantFiltration, bcftools and ANNOVAR 63 .
  • the variants in dbSNP, 1000 Genome 64 , EVS were filtered out.
  • the shared variants in four replicates of each group were then selected as the RNA editing sites.
  • the RNA editing level of Mock group was viewed as the background, and the global targets of Ctrl RNA 151 and arRNA 151 -PPIB were obtained by subtracting the variants in the Mock group.
  • X means the editing rate of each site in the Mock group
  • Y means the editing rate of each site in the Ctrl RNA 151 group (Fig. 6A) or arRNA 151 -PPIB group (Fig. 6B)
  • ⁇ x is the standard deviation of X
  • ⁇ Y is the standard deviation of Y
  • ⁇ X is the mean of X
  • ⁇ Y is the mean of Y
  • E is the expectation.
  • RNA-Seq data were analysed for the interrogation of possible transcriptional changes induced by RNA editing events.
  • the analysis of transcriptome-wide gene expression was performed using HISAT2 and STRINGTIE software 65 .
  • the sequencing reads were then mapped to reference genome (GRCh38-hg38) using HISAT2, followed by Pearson’s correlation coefficient analysis as mentioned above.
  • HEK293T cells were seeded on 12 wells plates (2 ⁇ 10 5 cells/well) . When approximately 70%confluent, cells were transfected with 1.5 ⁇ g of arRNA. As a positive control, 1 ⁇ g of poly (I: C) (Invitrogen, tlrl-picw) was transfected. Forty-eight hours later, cells were collected and subjected to RNA isolation (TIANGEN, DP430) . Then, the total RNAs were reverse-transcribed into cDNA via RT-PCR (TIANGEN, KR103-04) , and the expression of IFN- ⁇ and IL-6 were measured by quantitative PCR (TAKARA, RR820A) . The sequences of the primers were listed in the above table.
  • TP53 W53X cDNA-expressing plasmids and arRNA-expressing plasmids were co-transfected into HEK293T TP53 –/– cells, together with p53-Firefly-luciferase cis-reporting plasmids (YRGene, VXS0446) and Renilla-luciferase plasmids (a gift from Z. Jiang’s laboratory, Peking University) for detecting the transcriptional regulatory activity of p53. 48 hrs later, the cells were harvested and assayed with the Promega Dual-Glo Luciferase Assay System (Promega, E4030) according to the manufacturer protocol.
  • Dual-Glo Luciferase Reagent 150 ⁇ L Dual-Glo Luciferase Reagent was added to the harvested cell pellet, and 30 minutes later, the Firefly luminescence was measured by adding 100 ⁇ L Dual-Glo Luciferase Reagent (cell lysis) to 96-well white plate by Infinite M200 reader (TECAN) . 30 min later, 100 ⁇ L Dual-Glo stop and Glo Reagent were sequentially added to each well to measure the Renilla luminescence and calculate the ratio of Firefly luminescence to Renilla luminescence.
  • RNA oligo was dissolved in 100 ⁇ L opti-MEM medium (Gbico, 31985070) with the final concentration 2 ⁇ M. Then 1 ⁇ 10E6 GM06214 cells or 3 ⁇ 10E6 T cells were resuspended with the above electroporation mixture and electroporated with Agile Pulse In Vivo device (BTX) at 450 V for 1 ms. Then the cells were transferred to warm culture medium for the following assays.
  • BTX Agile Pulse In Vivo device
  • the harvested cell pellet was resuspended and lysed with 28 ⁇ L 0.5%Triton X-100 in 1 ⁇ PBS buffer on ice for 30 minutes. And then 25 ⁇ L of the cell lysis was added to 25 ⁇ L 190 ⁇ M 4-methylumbelliferyl- ⁇ -L-iduronidase substrate (Cayman, 2A-19543-500) , which was dissolved in 0.4 M sodium formate buffer containing 0.2%Triton X-100, pH 3.5, and incubated for 90 minutes at 37°C in the dark. The catalytic reaction was quenched by adding 200 ⁇ L 0.5M NaOH/Glycine buffer, pH 10.3, and then centrifuged for 2 minutes at 4°C. The supernatant was transferred to a 96-well plate, and fluorescence was measured at 365 nm excitation wavelength and 450 nm emission wavelength with Infinite M200 reader (TECAN) .
  • TECAN Infinite M200 reader
  • Example 1 Testing the RNA editing method of the invention on a reporter
  • Cas13 family proteins can edit RNA in mammalian cells.
  • C2c2 Cas13 family proteins
  • Dual fluorescence reporter-1 comprises sequence of mCherry (SEQ ID NO: 1) , sequence comprising 3 ⁇ GS linker and the targeted A (SEQ ID NO: 2) , and sequence of eGFP (SEQ ID NO: 3) .
  • Dual fluorescence reporter-2 comprises sequence of mCherry (SEQ ID NO: 1) , sequence comprising 3 ⁇ GS linker (shown as italic and bold characters) and the targeted A (shown as larger and bold A) (SEQ ID NO: 4) , and sequence of eGFP (SEQ ID NO: 3) .
  • Dual fluorescence reporter-3 comprises sequence of mCherry (SEQ ID NO: 1) , sequence comprising 1 ⁇ GS linker (shown as italic and bold characters) and the targeted A (SEQ ID NO: 5) , and sequence of eGFP (SEQ ID NO: 3) .
  • CTGCAGGGCGGAGGAGGCAGCGCCTGCTCGCGATGCTAGAGGGCTCTGCCA sequence comprising 1 ⁇ GS linker (shown as italic and bold characters) and the targeted A (shown as larger and bold A) ) (SEQ ID NO: 5)
  • the EGFP protein is substantially expressed.
  • the crRNA guide with the sequence: GGACCACCCCAAAAAUGAAUAUAACCAAAACUGAACAGCUCCUCGCCCUUGCUCACUGGCAGAGCCCUCCAGCAUCGCGAGCAGGCGCUGCCUCCUCCGCC(SEQ ID NO: 6) conferred over 25%EGFP positive efficiency. This indicates that adenine in the stop codon UAG is largely edited. In contrast, the random crRNA could not render the EGFP negative cells into positive (FIGs. 6A, 6B and 6C) . Based on these results, we inferred that overexpression of a RNA transcript alone could leverage endogenous ADAR enzyme to edit RNA.
  • RNA guides creating a linear guide RNA.
  • 70-nucleotides long RNA AAACCGAGGGAUCAUAGGGGACUGAAUCCACCAUUCUUCUCCCAAUCCCUGCAACUCCUUCUUCCCCUGC (SEQ ID NO: 7)
  • 70-nt random RNA UAAACAGCUCCUCGCCCUUGCUCACUGGCAGAGCCCUCCAGCAUCGCGAGCAGGCGCUGCCUCCUCCGCC
  • RNA Deaminase-recruiting RNA
  • Adar1 p110 and Adar1 p150 double knockout 293T cell lines FIGS. 6E and 6F.
  • Adar1 is ubiquitously expressed while Adar2 is mainly expressed in brain at high level. So we proposed the targeting Adenine deamination by dRNA was mainly mediated by Adar1 but not Adar2.
  • the targeting dRNA could not trigger EGFP expression in 293T-Adar1-/-cells, but overexpressing either exogenous Adar1 p110, p150 or Adar2 could rescue the EGFP expression in 293T-Adar1-/-cells (FIGs. 1E and 1F) , suggesting that in 293Tcells, the dRNA could recruit Adar1 or Adar2 to mediate adenine deamination on a target RNA.
  • Adar1-p110 and Adar2 have higher editing activity than Adar1-p150 (FIG. 1G and FIG. 6G) , possible due to the different cell localization of Adar1-p110 and Adar1-p150.
  • 16 different reporter comprising target sequences containing all possible 3 base motifs were constructed through Gibson cloning, and then cloned into pLenti backbone (pLenti-CMV-MCS-SV-Bsd, Stanley Cohen Lab, Stanford University) .
  • the target sequences are shown as follows.
  • dRNAs were kept same 111 bp length and designed a mismatch C at the center towards the target A.
  • dRNA could mediate mRNA transcribed from endogenous genes.
  • KRAS dRNA targeting four genes KRAS, PPIB, ⁇ -Actin and GAPDH.
  • KRAS mRNA we designed 91, 111, 131, 151, 171 and 191 nucleotides long dRNAs (FIG. 4A) with sequences as shown below.
  • the Next-generation sequencing results showed that the dRNAs could edit the targeted KRAS mRNA with up to 11.7%editing efficiency (FIG. 4B) .
  • the targeted three sites site1, site2 and site3.
  • We designed 151 nucleotides long dRNA for each site FIG. 4C with sequences as shown below.
  • the Next-generation sequencing results showed that the dRNA could edit PPIB mRNA site1 efficiently with up to 14%editing rate (FIG. 4D) .
  • the editing efficiency was 1.5%and 0.6% (FIG. 4E and 4F) .
  • endogenous ⁇ -Actin mRNA we selected two targeted site and designed dRNA for each site (FIG. 4G) with sequences as shown below.
  • dRNA could edit ⁇ -Actin mRNA both site1 and site2, with up to 1.4%editing efficiency for each site (FIG. 4H and FIG. 8A) .
  • longer dRNA conferred higher editing efficiency, with 0.6%for dRNA-71nt and 1.4%for dRNA-131nt (FIG. 3H) .
  • GAPDH housekeeping gene
  • flanking adenine could be edited (FIG. 5C and 5D) .
  • a variety of the off-targeted adenines were edited, while adenines such as A41, A43, A45, A46, A74, A79 showed more editing.
  • adenines such as A41, A43, A45, A46, A74, A79 showed more editing.
  • We found the 5’ nearest neighbor of those unedited A bases were G or C, whereas the 5’ nearest neighbor of those efficiently edited adenines was T or A.
  • ADAR preferred A-C mismatch to A-A, A-U, and, the A-G mismatch was the least preferred. So, we proposed that for the off-targeting A bases to which the 5’ nearest neighbor was U or A, A-G mismatch might reduce or diminish the off-targeting effects. Previous study has reported A-G mismatch could block the deamination editing by ADAR.
  • dRNA-AG1 A41, A46, A74
  • dRNA-AG2 A41, A43, A45, A46, A74, A79
  • dRNA-AG3 A31, A32, A33, A41, A43, A45, A46, A47, A74, A79
  • dRNA-AG4 A7, A31, A32, A33, A40, A41, A43, A45, A46, A47, A74, A79, A95) (FIG. 4E) .
  • dRNAs were transfected into HEK293T cells, and empty vector and 71-nt non-targeting dRNA control: (TCTCAGTCCAATGTATGGTCCGAGCACAAGCTCTAATCAAAGTCCGCGGGTGTAGACCGGTTGCCATAGGA (SEQ ID NO: 45) ) were used as negative controls.
  • the deep sequencing results showed that the on-target editing (A56) was reduced to 2.8%for dRNA-91-AG2, 2.3%for dRNA-91-AG3 and 0.7%for dRNA-91-AG4, compared to the on-target editing (A56) efficiency 7.9%for dRNA-91 without A-G mismatch (FIG. 4F) .
  • the on-target editing (A56) was reduced to 5.1%for dRNA-111-AG2 and 4.9%for dRNA-111-AG3 compared to the on-target editing (A56) efficiency 15.1%for dRNA-111 without A-G mismatch (FIG. 4F) , which indicating longer dRNA could bear more A-G mismatch.
  • 111-nt dRNA for detailed off-target analysis.
  • the flanking A bases editing were wiped out dramatically except for A7 and A79 (FIG. 4G) .
  • A7 base the off-target effect could be prevented by a further A-G mismatch design at this site, which is absent in the current dRNA design.
  • A79 base introducing adjacent two A-G mismatch A78/A79 might help to wipe out the off-target effects. Based on such results, applying the RNA editing systems of the present application to cure genetic diseases is very promising and encouraging.
  • Example 5 Testing an exemplary LEAPER method in multiple cell lines
  • RNA-guided RNA-targeting CRISPR effector 41 (Fig. 10A) .
  • surrogate reporter harbouring mCherry and EGFP genes linked by a sequence comprising a 3 ⁇ GGGGS-coding region and an in-frame UAG stop codon (Reporter-1, Fig. 10B) .
  • the reporter-transfected cells only expressed mCherry protein, while targeted editing on the UAG of the reporter transcript could convert the stop codon to UIG and consequently permit the downstream EGFP expression.
  • Such a reporter allows us to measure the A-to-I editing efficiency through monitoring EGFP level.
  • CRISPR RNAs hU6 promoter-driven crRNAs containing 5’ scaffolds subjected for Cas13a recognition and variable lengths of spacer sequences for targeting (crRNA Cas13a , following LbuCas13 crRNA sequences) .
  • the sequences complementary to the target transcripts all contain CCA opposite to the UAG codon so as to introduce a cytidine (C) mis-pairing with the adenosine (A) (Fig. 10B) because adenosine deamination preferentially occurs in the A-C mismatch site 13, 14 .
  • C cytidine
  • A adenosine
  • Fig. 10B adenosine deamination preferentially occurs in the A-C mismatch site 13, 14 .
  • non-targeting or targeting crRNAs of different lengths were co-expressed with dCas13a-ADAR1 DD proteins in HEK293T cells stably expressing the Reporter-1.
  • RNA editing effects indicated by the appearance of EGFP expression were observed with crRNAs containing matching sequences at least 40-nt long, and the longer the crRNAs the higher the EGFP positive percentage (Fig. 10C) .
  • Fig. 10C the longer the crRNAs the higher the EGFP positive percentage
  • Fig. 10C the co-expression of dCas13a-ADAR1 DD rather decreased crRNA activity.
  • the EGFP expression was clearly sequence-dependent because the 70-nt (exclusive of the 5’ scaffold for the length calculation) control RNA could not activate EGFP expression (Figs. 10C, 10D) .
  • Example 7 LEAPER enables RNA editing in multiple cell lines
  • ADAR1 was highly expressed in all tested cell lines, and its identity in the Western blots was confirmed by the negative control, HEK293T ADAR1 –/– line (Fig. 14A, B) .
  • ADAR3 was detected only in HepG2 and HeLa cells (Fig. 14A, B) .
  • ADAR2 was non-detectable in any cells, a result that was not due to the failure of Western blotting because ADAR2 protein could be detected from ADAR2-overexpressing HEK293T cells (Fig. 14A, B) . These findings are in consistent with previous reports that ADAR1 is ubiquitously expressed, while the expressions of ADAR2 and ADAR3 are restricted to certain tissues 11 .
  • LEAPER worked in all tested cells for this arRNA 71 , albeit with varying efficiencies (Fig. 14C) . These results were in agreement with the prior report that the ADAR1/2 protein levels correlate with the RNA editing yield 42 , with the exception of HepG2 and HeLa cells. The suboptimal correlations of editing efficiencies with ADAR1 levels were likely due to the abundant ADAR3 expressions in these two lines (Fig. 14A, B) because it has been reported that ADAR3 plays an inhibitory role in RNA editing. Importantly, LEAPER also worked in three different cell lines of mouse origin (NIH3T3, Mouse Embryonic Fibroblast (MEF) and B16) (Fig. 14D) , paving the way for testing its therapeutics potential through animal and disease models. Collectively, we conclude that LEAPER is a versatile tool for wide-spectrum of cell types, and for different organisms.
  • RNA Length of RNA appeared relevant to arRNA efficiency in directing the editing on the targeted transcripts (Fig. 10C) , consistent with a previous report 42 .
  • arRNAs with variable lengths targeting two different reporter transcripts-Reporter-1 and Reporter-2 (Fig. 15A, B) .
  • arRNAs of 10 different sizes were designed and tested, ranging from 31-nt to 211-nt, with CCA triplet (for UAG targeting) right in the middle (Sequences of arRNAs and control RNAs used in this study listed above) .
  • the length of arRNA correlated positively with the editing efficiency, for both reporters, peaking at 111-to191-nt (Fig. 16D) .
  • 71-nt was the minimal length for arRNA to work for both reporters (Fig. 16D) .
  • the 151-nt arRNA PPIB edited ⁇ 50%of total transcripts of PPIB gene (Fig. 17B) .
  • No arRNAs showed RNAi effects on their targeted transcripts (Fig. 18A) or ultimate protein level (e.g. KRAS, Fig. 18B) .
  • LEAPER is able to achieve desirable editing rate on non-UAN sites (Fig. 17C and Sequences of arRNAs and control RNAs used in this study listed above) , showing the flexibility of LEAPER on editing endogenous transcripts. To further explore the power of LEAPER, we tested whether it could simultaneously target multiple sites.
  • ADAR1/2 tend to promiscuously deaminate multiple adenosines in an RNA duplex 44 and the A-C mismatch is not the only motif to guide the A-to-I switch (Fig. 16A) . It is therefore reasonable to assume that all adenosines on target transcripts within the arRNA coverages are subjected to variable levels of editing, major sources of unwanted modifications. The longer the arRNA, the higher the possibility of such off-targets. We therefore examined all adenosine sites within the arRNA covering regions in these targeted transcripts. For PPIB transcripts, very little off-target editing was observed throughout the sequencing window for variable sizes of arRNAs (Fig. 17E, F) .
  • arRNA 111 -AG6 we then added more mismatches on arRNA 111 -AG6, including a dual mismatch (5’ -CGG opposite to the targeted motif5’ -AAG) , plus three additional A-G mismatches to mitigate editing on the 27 th , 98 th and the 115 th adenosines (arRNA 111 -AG9) (Sequences of arRNAs and control RNAs used in this study listed above) . Consequently, we achieved a much improved specificity for editing, without additional loss of editing rate on the targeted site (A76) (Fig. 17I) .
  • engineered LEAPER incorporating additional rules enables efficient and more precise RNA editing on endogenous transcripts.
  • RNA duplex With the target transcript and that RNA duplex might elicit innate immune response, we investigated if the introduction of arRNA has such an effect. To test this, we selected arRNAs targeting four gene transcripts that had been proven effective. We did not observe any mRNA induction of interferon- ⁇ (IFN- ⁇ ) (Fig. 22E) or interleukin-6 (IL-6) (Fig. 22F) , which are two hallmarks of innate immune activation. As a positive control, a synthetic analog of double-stranded RNA-poly (I: C) induced strong IFN- ⁇ and IL-6 expression (Fig. 22E, F) . LEAPER does not seem to induce immunogenicity in target cells, a feature important for safe therapeutics.
  • IFN- ⁇ interferon- ⁇
  • IL-6 interleukin-6
  • TP53 tumor suppressor gene
  • LEAPER could be used to correct more pathogenic mutations.
  • Aiming at clinically relevant mutations from six pathogenic genes, COL3A1 of Ehlers-Danlos syndrome, BMPR2 of Primary pulmonary hypertension, AHI1 of Joubert syndrome, FANCC of Fanconi anemia, MYBPC3 of Primary familial hypertrophic cardiomyopathy and IL2RG of X-linked severe combined immunodeficiency we designed 111-nt arRNAs for each of these genes carrying corresponding pathogenic G>A mutations (Fig. 25 and Sequences of arRNAs and control RNAs used in this study listed above, and the following disease-relevant cDNAs used in this study) .
  • arRNA oligonucleotides were chemically synthesized with 2’ -O-methyl and phosphorothioate linkage modifications at the first three and last three residues of arRNAs (Fig. 28C) .
  • arRNA 111 -PPIB oligos achieved ⁇ 20%of editing on PPIB transcripts (Fig. 28D) , indicating that LEAPER holds promise for the development of oligonucleotide drugs.
  • Example 16 Restoration of ⁇ -L-iduronidase activity in Hurler syndrome patient-derived primary fibroblast by LEAPER
  • Adeno-associated Virus (AAV) is selected as the delivery system.
  • the two sequences (Seq ID NO. 341 and Seq ID NO. 342) were inserted into the plasmid AAV-U6-CMV-GFP vector, and the plasmid was packaged into AAV8 virus by PackGene Biotech (see FIG. 31) .
  • the virus titer was as follows:
  • mice Six MPSI (idua W392X mice, B6.129S-Idua tm1.1Kmke/J ) male mice (4-10 weeks old) were selected and divided into two groups, and each of three mice of the first group was injected with 1E12 GC of AAV8 virus packed with plasmid AAV8-IDUA-adRNA151-KD2 through the tail vein, each of other three mice of the second group were injected with 1E12 GC of AAV8 virus packed with plasmid AAV8-Random151-KD2 through the tail vein as well as a control. Twenty-eight days after the injection, the mice were sacrificed by cervical dislocation and livers were taken. .
  • Coding sequence (CDS) of the disease-relevant genes
  • Genome editing technologies are revolutionizing biomedical research.
  • Highly active nucleases such as zinc finger nucleases (ZFNs) 1 , transcription activator-like effector nucleases (TALENs) 2-4 , and Cas proteins of CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) system 5-7 have been successfully engineered to manipulate the genome in a myriad of organisms.
  • ZFNs zinc finger nucleases
  • TALENs transcription activator-like effector nucleases
  • Cas proteins of CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • RNA is an attractive target for genetic correction because RNA modification could alter the protein function without generating any permanent changes to the genome.
  • the ADAR adenosine deaminases are currently exploited to achieve precise base editing on RNAs.
  • Three kinds of ADAR proteins have been identified in mammals, ADAR1 (isoforms p110 and p150) , ADAR2 and ADAR3 (catalytic inactive) 11, 12 , whose substrates are double-stranded RNAs, in which an adenosine (A) mismatched with a cytosine (C) is preferentially deaminated to inosine (I) .
  • Inosine is believed to mimic guanosine (G) during translation 13, 14 .
  • the ADAR protein or its catalytic domain was fused with a ⁇ N peptide 15-17 , a SNAP-tag 18-22 or a Cas protein (dCas13b) 23 , and a guide RNA was designed to recruit the chimeric ADAR protein to the specific site.
  • a guide RNA was designed to recruit the chimeric ADAR protein to the specific site.
  • overexpressing ADAR1 or ADAR2 proteins together with an R/G motif-bearing guide RNA was also reported to enable targeted RNA editing 24-27 .
  • ADAR1 adeno-associated virus
  • ectopic expression of proteins or their domains of non-human origin has potential risk of eliciting immunogenicity 30, 33 .
  • pre-existing adaptive immunity and p53-mediated DNA damage response may compromise the efficacy of the therapeutic protein, such as Cas9 34-38 .
  • endogenous mechanism for RNA editing this was tried only by injecting pre-assembled target transcript: RNA duplex into Xenopus embryos 39 .
  • Alternative technologies for robust nucleic acid editing that don’t rely on ectopic expression of proteins are much needed.
  • expressing a deliberately designed guide RNA enables efficient and precise editing on endogenous RNAs, and corrects pathogenic mutations. This unary nucleic acid editing platform may open new avenues for therapeutics and research.
  • LEAPER catalyzes the precise A to I switch without generating cutting or degradation of targeted transcripts (Fig. 18A) .
  • Fig. 22E, f and Fig. 29E the length requirement for arRNA is longer than RNAi, it neither induces immune-stimulatory effects at the cellular level (Fig. 22E, f and Fig. 29E) nor affects the function of endogenous ADAR proteins (Fig. 22A, B) ,making it a safe strategy for RNA targeting.
  • ectopic expression of ADAR proteins or their catalytic domains induces substantial global off-target edits 32 and possibly triggers cancer 31 .
  • LEAPER cytosine base editor could generate substantial off-target single-nucleotide variants in mouse embryos, rice or human cell lines due to the expression of an effector protein, which illustrates the advantage of LEAPER for potential therapeutic application 52-54 .
  • LEAPER empowers efficient editing while elicits rare global off-target editing (Fig. 20 and Fig. 21) .
  • LEAPER could minimize potential immunogenicity or surmount delivery obstacles commonly shared by other methods that require the introduction of foreign proteins.
  • RNA duplex longer than 70-nt is stoichiometrically important for recruiting or docking ADAR proteins for effective editing.
  • longer arRNA resulted in higher editing yield in both ectopically expressed reporters and endogenous transcripts (Fig. 16D and Fig. 17B) .
  • ADAR proteins promiscuously deaminate adenosine base in the RNA duplex, longer arRNA may incur more off-targets within the targeting window.
  • LEAPER could effectively target native transcripts, their editing efficiencies and off-target rates varied.
  • PPIB transcript-targeting we could convert 50%of targeted adenosine to inosine without evident off-targets within the covering windows (Fig. 17B, F) .
  • the off-targets became more severe for other transcripts.
  • We have managed to reduce off-targets such as introducing A-G mismatches or consecutive mismatches to repress undesired editing.
  • too many mismatches could decrease on-target efficiency.
  • LEAPER could be harnessed to manipulate gene function or correct pathogenic mutation.
  • LEAPER is not limited to only work on UAG, instead that it works with possibly any adenosine regardless of its flanking nucleotides (Fig. 16F, G and Fig. 17C) .
  • Such flexibility is advantageous for potential therapeutic correction of genetic diseases caused by certain single point mutations.
  • the arRNA targeting pre-mRNA is more effective than that targeting mature RNA, indicating that nuclei are the main sites of action for ADAR proteins and LEAPER could be leveraged to manipulate splicing by modifying splice sites within pre-mRNAs. What’s more, LEAPER has demonstrated high efficiency for simultaneously targeting multiple gene transcripts (Fig. 17D) . This multiplexing capability of LEAPER might be developed to cure certain polygenetic diseases in the future.
  • RNA editing approaches safer for therapeutics than means of genome editing.
  • transient editing is well suited for temporal control of treating diseases caused by occasional changes in a specific state.
  • LEAPER and other RNA editing methods would not introduce DSB on the genome, avoiding the risk of generating undesirable deletions of large DNA fragments 37 .
  • DNA base editing methods adopting nickase Cas9 could still generate indels in the genome 8 .
  • LEAPER should also work in post-mitosis cells such as cerebellum cells with high expression ofADAR2 11 .
  • LEAPER could apply to a broad spectrum of cell types such as human cell lines (Fig. 14C) , mouse cell lines (Fig. 14D) and human primary cells including primary T cells (Fig. 27 and Fig. 28D) . Efficient editing through lentiviral delivery or synthesized oligo provides increased potential for therapeutic development (Fig. 28) . Moreover, LEAPER could produce phenotypic or physiological changes in varieties of applications including recovering the transcriptional regulatory activity of p53 (Fig. 7) , correcting pathogenic mutations (Fig. 26) , and restoring the ⁇ -L-iduronidase activity in Hurler syndrome patient-derived primary fibroblasts (Fig. 29) . It can thus be envisaged that LEAPER has enormous potential in disease treatment.
  • RESTORE RNA editing method
  • the fundamental difference between RESTORE and LEAPER lies in the distinct nature of the guide RNA for recruiting endogenous ADAR.
  • the guide RNA of RESTORE is limited to chemosynthetic antisense oligonucleotides (ASO) depending on complex chemical modification, while arRNA of LEAPER can be generated in a variety of ways, chemical synthesis and expression from viral or non-viral vectors (Fig. 28 and Fig. 29) .
  • ASOs is restricted to act transiently in disease treatment.
  • arRNA could be produced through expression, a feature particularly important for the purpose of constant editing.
  • LEAPER relies on the endogenous ADAR
  • the expression level of ADAR proteins in target cells is one of the determinants for successful editing.
  • the ADAR1 p110 is ubiquitously expressed across tissues, assuring the broad applicability of LEAPER.
  • the ADAR1 p150 is an interferon-inducible isoform 58 , and has proven to be functional in LEAPER (Fig. 11E, Fig. 12B) .
  • co-transfection of interferon stimulatory RNAs with the arRNA might further improve editing efficiency under certain circumstances.
  • ADAR3 plays inhibitory roles
  • inhibition of ADAR3 might enhance editing efficiency in ADAR3-expressing cells.
  • additional modification of arRNA might increase its editing efficiency.
  • arRNA fused with certain ADAR-recruiting scaffold may increase local ADAR protein concentration and consequently boost editing yield. So far, we could only leverage endogenous ADAR1/2 proteins for the A to I base conversion. It is exciting to explore whether more native mechanisms could be harnessed similarly for the modification of genetic elements, especially to realize potent nucleic acid editing.
  • LEAPER is a simple, efficient and safe system, shedding light on a novel path for gene editing-based therapeutics and research.
  • Nishikura, K Functions and regulation of RNA editing by ADAR deaminases. Annu Rev Biochem 79, 321-349 (2010) .
  • RNA 7, 846-858 (2001) .
  • RNA-specific adenosine deaminase gene family contains both single-and double-stranded RNA binding domains. RNA 6, 755-767 (2000) .
  • Double-stranded RNA unwinding and modifying activity is detected ubiquitously in primary tissues and cell lines. Mol Cell Biol 10, 5586-5590 (1990) .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Cardiology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Diabetes (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
PCT/CN2019/110782 2018-10-12 2019-10-12 Methods and Compositions for Editing RNAs WO2020074001A1 (en)

Priority Applications (26)

Application Number Priority Date Filing Date Title
KR1020217014349A KR102666695B1 (ko) 2018-10-12 2019-10-12 Rna를 편집하기 위한 방법 및 조성물
EA202191004A EA202191004A1 (ru) 2019-04-15 2019-10-12 Способы и композиции для редактирования рнк
AU2019357450A AU2019357450B2 (en) 2018-10-12 2019-10-12 Methods and compositions for editing RNAs
CA3115864A CA3115864A1 (en) 2018-10-12 2019-10-12 Methods and compositions for editing rnas
EP19870299.5A EP3864152B1 (en) 2018-10-12 2019-10-12 Methods and compositions for editing rnas
EP23184286.5A EP4306116A3 (en) 2018-10-12 2019-10-12 Methods and compositions for editing rnas
PE2021000498A PE20211402A1 (es) 2018-10-12 2019-10-12 Metodos y composiciones para editar acidos ribonucleicos
JP2021520227A JP7252328B2 (ja) 2018-10-12 2019-10-12 Rnaを編集する方法および組成物
CN202210724157.3A CN116218836A (zh) 2018-10-12 2019-10-12 编辑rna的方法和组合物
US17/284,232 US20210355494A1 (en) 2018-10-12 2019-10-12 Methods and compositions for editing rnas
SG11202103666PA SG11202103666PA (en) 2018-10-12 2019-10-12 Methods and Compositions for Editing RNAs
ES19870299T ES2962434T3 (es) 2018-10-12 2019-10-12 Procedimientos y composiciones para editar ARN
MX2021004187A MX2021004187A (es) 2018-10-12 2019-10-12 Métodos y composiciones para editar ácidos ribonucleicos.
BR112021006844A BR112021006844A8 (pt) 2018-10-12 2019-10-12 Métodos para editar um rna alvo e para tratar ou prevenir uma doença ou condição, rna editado ou uma célula hospedeira tendo um rna editado, rna que recruta desaminase, construção, biblioteca, composição, célula hospedeira, e, kit para editar um rna alvo .
CR20210243A CR20210243A (es) 2018-10-12 2019-10-12 Métodos y composiciones para editar ácidos ribonucleicos
CN202210740517.9A CN116042611A (zh) 2018-10-12 2019-10-12 编辑rna的方法和组合物
CN201980067380.1A CN113631708B (zh) 2018-10-12 2019-10-12 编辑rna的方法和组合物
CN202210717093.4A CN115651927B (zh) 2018-10-12 2019-10-12 编辑rna的方法和组合物
DK19870299.5T DK3864152T3 (da) 2018-10-12 2019-10-12 Fremgangsmåder og sammensætninger til editering af rna'er
IL282191A IL282191A (en) 2018-10-12 2021-04-08 Methods and preparations for editing RN" A
US17/226,961 US20210310026A1 (en) 2018-10-12 2021-04-09 Methods and compositions for editing rnas
PH12021550805A PH12021550805A1 (en) 2018-10-12 2021-04-12 Methods and Compositions for Editing RNAs
CONC2021/0005956A CO2021005956A2 (es) 2018-10-12 2021-05-06 Métodos y composiciones para editar ácidos ribonucleicos
ECSENADI202133637A ECSP21033637A (es) 2018-10-12 2021-05-12 Methods and Compositions for Editing RNAs
JP2023045897A JP2023078373A (ja) 2018-10-12 2023-03-22 Rnaを編集する方法および組成物
AU2023202184A AU2023202184A1 (en) 2018-10-12 2023-04-11 Methods and Compositions for Editing RNAs

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CNPCT/CN2018/110105 2018-10-12
CN2018110105 2018-10-12
CNPCT/CN2019/082713 2019-04-15
CN2019082713 2019-04-15

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/226,961 Continuation US20210310026A1 (en) 2018-10-12 2021-04-09 Methods and compositions for editing rnas

Publications (1)

Publication Number Publication Date
WO2020074001A1 true WO2020074001A1 (en) 2020-04-16

Family

ID=70163607

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/110782 WO2020074001A1 (en) 2018-10-12 2019-10-12 Methods and Compositions for Editing RNAs

Country Status (21)

Country Link
US (2) US20210355494A1 (es)
EP (2) EP4306116A3 (es)
JP (2) JP7252328B2 (es)
KR (1) KR102666695B1 (es)
CN (4) CN115651927B (es)
AU (2) AU2019357450B2 (es)
BR (1) BR112021006844A8 (es)
CA (1) CA3115864A1 (es)
CL (1) CL2021000880A1 (es)
CO (1) CO2021005956A2 (es)
CR (1) CR20210243A (es)
DK (1) DK3864152T3 (es)
EC (1) ECSP21033637A (es)
ES (1) ES2962434T3 (es)
IL (1) IL282191A (es)
MX (1) MX2021004187A (es)
PE (1) PE20211402A1 (es)
PH (1) PH12021550805A1 (es)
SG (1) SG11202103666PA (es)
TW (1) TW202028466A (es)
WO (1) WO2020074001A1 (es)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021136404A1 (zh) 2019-12-30 2021-07-08 博雅辑因(北京)生物科技有限公司 一种治疗Usher综合征的方法和其组合物
WO2021136408A1 (zh) 2019-12-30 2021-07-08 博雅辑因(北京)生物科技有限公司 一种基于leaper技术治疗mps ih的方法和组合物
WO2021209010A1 (zh) * 2020-04-15 2021-10-21 博雅辑因(北京)生物科技有限公司 一种治疗赫勒氏综合征的方法和药物
WO2021243023A1 (en) * 2020-05-28 2021-12-02 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of serpina1
CN113897359A (zh) * 2020-07-06 2022-01-07 博雅辑因(北京)生物科技有限公司 一种改善的rna编辑方法
WO2022007803A1 (zh) 2020-07-06 2022-01-13 博雅辑因(北京)生物科技有限公司 一种改善的rna编辑方法
WO2022091100A1 (en) 2020-10-29 2022-05-05 Yeda Research And Development Co. Ltd. Polynucleotides for rna editing and methods of using same
WO2022147573A1 (en) * 2021-01-04 2022-07-07 The Regents Of The University Of California Programmable rna editing in vivo via recruitment of endogenous adars
US11479775B2 (en) 2017-03-03 2022-10-25 The Regents Of The University Of California RNA targeting of mutations via suppressor tRNAs and deaminases
WO2023278436A1 (en) * 2021-06-29 2023-01-05 Shape Therapeutics Inc. Engineered guide rnas and polynucleotides
WO2023020574A1 (en) * 2021-08-18 2023-02-23 Peking University Engineered adar-recruiting rnas and methods of use thereof
WO2023077135A1 (en) * 2021-10-29 2023-05-04 Duke University Compositions and systems for rna-programmable cell editing and methods of making and using same
US11661596B2 (en) 2019-07-12 2023-05-30 Peking University Targeted RNA editing by leveraging endogenous ADAR using engineered RNAs
US11702658B2 (en) 2019-04-15 2023-07-18 Edigene Therapeutics (Beijing) Inc. Methods and compositions for editing RNAs
US11827880B2 (en) 2019-12-02 2023-11-28 Shape Therapeutics Inc. Therapeutic editing
WO2024086650A1 (en) * 2022-10-18 2024-04-25 Locanabio, Inc. Compositions and methods comprising programmable snrnas for rna editing

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117015605A (zh) * 2021-01-12 2023-11-07 北京大学 使用工程化rna通过利用内源性adar进行靶向rna编辑
US20230123513A1 (en) * 2021-06-15 2023-04-20 Massachusetts Institute Of Technology Deaminase-based rna sensors
WO2023060186A1 (en) 2021-10-07 2023-04-13 Hepatx Corporation Methods of tracking donor cells in a recipient
WO2023143539A1 (en) * 2022-01-28 2023-08-03 Edigene Therapeutics (Beijing) Inc. Engineered adar-recruiting rnas and methods of use thereof
WO2023185231A1 (en) * 2022-04-02 2023-10-05 Edigene Therapeutics (Beijing) Inc. Engineered adar-recruiting rnas and methods of use for usher syndrome

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016097212A1 (en) * 2014-12-17 2016-06-23 Proqr Therapeutics Ii B.V. Targeted rna editing
WO2017220751A1 (en) * 2016-06-22 2017-12-28 Proqr Therapeutics Ii B.V. Single-stranded rna-editing oligonucleotides
WO2018041973A1 (en) * 2016-09-01 2018-03-08 Proqr Therapeutics Ii B.V. Chemically modified single-stranded rna-editing oligonucleotides
WO2018161032A1 (en) * 2017-03-03 2018-09-07 The Regents Of The University Of California RNA TARGETING OF MUTATIONS VIA SUPPRESSOR tRNAs AND DEAMINASES

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101297830B1 (ko) * 2010-09-10 2013-08-19 한국생명공학연구원 신규한 rna 결합 단백질 및 이를 이용한 무표지 마이크로 rna 검출방법
EP3230445B1 (en) * 2014-12-12 2024-01-24 Tod M. Woolf Compositions and methods for editing nucleic acids in cells utilizing oligonucleotides
US11390865B2 (en) * 2015-07-14 2022-07-19 Fukuoka University Method for introducing site-directed RNA mutation, target editing guide RNA used in the method and target RNA-target editing guide RNA complex
DE102015012522B3 (de) * 2015-09-26 2016-06-02 Eberhard Karls Universität Tübingen Verfahren und Substanzen zur gerichteten RNA-Editierung
WO2018134301A1 (en) * 2017-01-19 2018-07-26 Proqr Therapeutics Ii B.V. Oligonucleotide complexes for use in rna editing
KR102185464B1 (ko) * 2017-03-15 2020-12-03 매사추세츠 인스티튜트 오브 테크놀로지 신규 cas13b 오르소로그 crispr 효소 및 시스템

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016097212A1 (en) * 2014-12-17 2016-06-23 Proqr Therapeutics Ii B.V. Targeted rna editing
WO2017220751A1 (en) * 2016-06-22 2017-12-28 Proqr Therapeutics Ii B.V. Single-stranded rna-editing oligonucleotides
WO2018041973A1 (en) * 2016-09-01 2018-03-08 Proqr Therapeutics Ii B.V. Chemically modified single-stranded rna-editing oligonucleotides
WO2018161032A1 (en) * 2017-03-03 2018-09-07 The Regents Of The University Of California RNA TARGETING OF MUTATIONS VIA SUPPRESSOR tRNAs AND DEAMINASES

Non-Patent Citations (72)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1980
ABUDAYYEH, O. O. ET AL.: "C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector", SCIENCE, vol. 353, 2016, pages aaf5573, XP055407082, DOI: 10.1126/science.aaf5573
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1999, JOHN WILEY & SONS
BASS, B. L.WEINTRAUB, H.: "An unwinding activity that covalently modifies its double-stranded RNA substrate", CELL, vol. 55, 1988, pages 1089 - 1098, XP024244880, DOI: 10.1016/0092-8674(88)90253-X
BOCH, J.: " Breaking the code of DNA binding specificity of TAL-type III effectors", SCIENCE, vol. 326, 2009, pages 1509 - 1512, XP055250971, DOI: 10.1126/science.1178811
CHARLESWORTH, C. T. ET AL.: "Identification of preexisting adaptive immunity to Cas9 proteins in humans", NATMED, vol. 25, 2019, pages 249 - 254, XP036693195, DOI: 10.1038/s41591-018-0326-x
CHEN, C. X. ET AL.: "A third member of the RNA-specific adenosine deaminase gene family, ADAR3, contains both single- and double-stranded RNA binding domains", RNA, vol. 6, 2000, pages 755 - 767
CHEW, W. L. ET AL.: "A multifunctional AAV-CRISPR-Cas9 and its host response", NAT METHODS, vol. 13, 2016, pages 868 - 874, XP055339896, DOI: 10.1038/nmeth.3993
CONG, L. ET AL.: "Multiplex genome engineering using CRISPR/Cas systems", SCIENCE, vol. 339, 2013, pages 819 - 823, XP055400719, DOI: 10.1126/science.1231143
COX, D. B. T. ET AL.: "RNA editing with CRISPR-Cas13", SCIENCE, vol. 358, 2017, pages 1019 - 1027, XP055491658, DOI: 10.1126/science.aaq0180
COX, D.B.T. ET AL.: "RNA editing with CRISPR-Casl3", SCIENCE, vol. 358, 24 November 2017 (2017-11-24), pages 1019 - 1027, XP055491658, DOI: 10.1126/science.aaq0180 *
DANIEL, C., WIDMARK, A., RIGARDT, D., OHMAN, M.: "Editing inducer elements increases A-to-l editing efficiency in the mammalian transcriptome", GENOME BIOL, vol. 18, 2017, pages 195, XP055809499, DOI: 10.1186/s13059-017-1324-x
DOBIN, A. ET AL.: "STAR: ultrafast universal RNA-seq aligner", BIOINFORMATICS, vol. 29, 2013, pages 15 - 21, XP055500895, DOI: 10.1093/bioinformatics/bts635
DOUBROVIN, M. ET AL.: "Imaging transcriptional regulation of p53-dependent genes with positron emission tomography in vivo", PROC NATL ACAD SCI U S A, vol. 98, 2001, pages 9300 - 9305, XP002285743, DOI: 10.1073/pnas.161091198
FIRE, A. ET AL.: "Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans", NATURE, vol. 391, 1998, pages 806 - 811, XP002199982, DOI: 10.1038/35888
FLOQUET, C.DEFORGES, J.ROUSSET, J. P.BIDOU, L.: "Rescue of non-sense mutated p53 tumor suppressor gene by aminoglycosides", NUCLEIC ACIDS RES, vol. 39, 2011, pages 3350 - 3362, XP055370024, DOI: 10.1093/nar/gkq1277
FUKUDA, M. ET AL.: "Construction of a guide-RNA for site-directed RNA mutagenesis utilising intracellular A-to-I RNA editing", SCIENTIFIC REPORTS, vol. 7, 2017, pages 41478
GAUDELLI, N. M. ET AL.: "Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage", NATURE, vol. 551, 2017, pages 464 - 471
GENOMES PROJECT, C. ET AL.: "An integrated map of genetic variation from 1,092 human genomes", NATURE, vol. 491, 2012, pages 56 - 65, XP055144425, DOI: 10.1038/nature11632
GIBSON, D. G. ET AL.: "Enzymatic assembly of DNA molecules up to several hundred kilobases", NAT METHODS, vol. 6, 2009, pages 343 - 345, XP055224105, DOI: 10.1038/nmeth.1318
HAAPANIEMI, E.BOTLA, S.PERSSON, J.SCHMIERER, B.TAIPALE, J.: "CR!SPR-Cas9 genome editing induces a p53-mediated DNA damage response", NAT MED, vol. 24, 2018, pages 927 - 930, XP036542072, DOI: 10.1038/s41591-018-0049-z
HANSWILLEMENKE, A.KUZDERE, T.VOGEL, P.JEKELY, G.STAFFORST, T.: "Site-Directed RNA Editing in Vivo Can Be Triggered by the Light-Driven Assembly of an Artificial Riboprotein", JAM CHEM SOC, vol. 137, 2015, pages 15875 - 15881, XP055558873, DOI: 10.1021/jacs.5b10216
HEEP, M.MACH, P.REAUTSCHNIG, P.WETTENGEL, J.STAFFORST, T.: "Applying Human ADAR1p110 and ADAR1p150 for Site-Directed RNA Editing-G/C Substitution Stabilizes GuideRNAs against Editing", GENES (BASEL), vol. 8, 2017, XP055586052, DOI: 10.3390/genes8010034
IHRY, R. J. ET AL.: "p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells", NATMED, vol. 24, 2018, pages 939 - 946, XP036542073, DOI: 10.1038/s41591-018-0050-6
JIN, S. ET AL.: "Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice", SCIENCE, 2019
JINEK, M. ET AL.: "A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity", SCIENCE, vol. 337, 2012, pages 816 - 821, XP055229606, DOI: 10.1126/science.1225829
KATREKAR, D. ET AL.: "In vivo RNA editing of point mutations via RNA-guided adenosine deaminases", NAT METHODS, vol. 16, 2019, pages 239 - 242, XP036713087, DOI: 10.1038/s41592-019-0323-0
KERN, S. E.: "Identification of p53 as a sequence-specific DNA-binding protein", SCIENCE, vol. 252, 1991, pages 1708 - 1711
KIM, D., KIM, D. E., LEE, G., CHO, S. I., KIM, J. S.: "Genome-wide target specificity of CRISPR RNA-guided adenine base editors", NAT BIOTECHNOL, vol. 37, 2019, pages 430 - 435, XP036900639, DOI: 10.1038/s41587-019-0050-1
KOMOR, A. C.KIM, Y. B.PACKER, M. S.ZURIS, J. A.LIU, D. R.: "Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage", NATURE, vol. 533, 2016, pages 420 - 424, XP055968803, DOI: 10.1038/nature17946
LANDRUM, M. J. ET AL.: "ClinVar: public archive of interpretations of clinically relevant variants", NUCLEIC ACIDS RES, vol. 44, 2016, pages D862 - 868, XP055715955, DOI: 10.1093/nar/gkv1222
LEVANON, E. Y. ET AL.: "Systematic identification of abundant A-to-I editing sites in the human transcriptome", NAT BIOTECHNOL, vol. 22, 2004, pages 1001 - 1005, XP002336893, DOI: 10.1038/nbt996
MA, Y. ET AL.: "Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells", NAT METHODS, vol. 13, 2016, pages 1029 - 1035, XP055573969, DOI: 10.1038/nmeth.4027
MALI, P. ET AL.: "RNA-guided human genome engineering via Cas9", SCIENCE, vol. 339, 2013, pages 823 - 826, XP055469277, DOI: 10.1126/science.1232033
MAYS, L. E.WILSON, J. M.: "The complex and evolving story of T cell activation to AAV vector-encoded transgene products", MOL THER, vol. 19, 2011, pages 16 - 27
MERKLE, T. ET AL.: "Precise RNA editing by recruiting endogenous ADARs with antisense oligonucleotides", NATBIOTECHNOL, vol. 37, 2019, pages 133 - 138, XP036900581, DOI: 10.1038/s41587-019-0013-6
MILLER, J. C. ET AL.: "A TALE nuclease architecture for efficient genome editing", NAT, vol. 29, 2011, pages 143 - 148, XP055568321, DOI: 10.1038/nbt.1755
MONTIEL-GONZALEZ, M. F.VALLECILLO-VIEJO, I. C.ROSENTHAL, J. J.: "An efficient system for selectively altering genetic information within mRNAs", NUCLEIC ACIDS RES, vol. 44, 2016, pages e157
MONTIEL-GONZALEZ, M. F.VALLECILLO-VIEJO, I.YUDOWSKI, G. A.ROSENTHAL, J. J.: "Correction of mutations within the cystic fibrosis transmembrane conductance regulator by site-directed RNA editing", PROC NATL ACAD SCI U S A, vol. 110, 2013, pages 18285 - 18290, XP055260494, DOI: 10.1073/pnas.1306243110
MOSCOU, M. J.BOGDANOVE, A. J.: "A simple cipher governs DNA recognition by TAL effectors", SCIENCE, vol. 326, 2009, pages 1501, XP002599998
NISHIKURA, K.: "Functions and regulation of RNA editing by ADAR deaminases", ANNU REV BIOCHEM, vol. 79, 2010, pages 321 - 349, XP055800809
NISHIKURA, K: "A-to-I editing of coding and non-coding RNAs by ADARs", NAT REV MOL, 2016, pages 83 - 96
OU, L.: " ZFN-Mediated In Vivo Genome Editing Corrects Murine Hurler Syndrome", MOL THER, vol. 27, 2019, pages 178 - 187
PATTERSON, J. B.SAMUEL, C. E.: "Expression and regulation by interferon of a double-stranded-RNA-specific adenosine deaminase from human cells: evidence for two forms of the deaminase", MOL CELL BIOL, vol. 15, 1995, pages 5376 - 5388, XP002215980
PERTEA, M.KIM, D.PERTEA, G. M.LEEK, J. T.SALZBERG, S. L.: "Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown", NAT PROTOC, vol. 11, 2016, pages 1650 - 1667, XP055596735, DOI: 10.1038/nprot.2016.095
PLATT, R. J.: "CRISPR-Cas9 knockin mice for genome editing and cancer modeling", CELL, vol. 159, 2014, pages 440 - 455, XP055523070, DOI: 10.1016/j.cell.2014.09.014
PORTEUS, M. H.CARROLL, D.: "Gene targeting using zinc finger nucleases", NAT BIOTECHNOL, vol. 23, 2005, pages 967 - 973, XP002467422, DOI: 10.1038/nbt1125
QU, L. ET AL.: "Programmable RNA editing by recruiting endogenous ADAR using engineered RNAs", NATURE BIOTECHNOLOGY, vol. 37, 30 September 2019 (2019-09-30), pages 1059 - 1069, XP036888288, DOI: 10.1038/s41587-019-0178-z *
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR PRESS
SAVVA, Y. A.RIEDER, L. E.REENAN, R. A.: "The ADAR protein family", GENOME BIOL, vol. 13, 2012, pages 252
SCHNEIDER, M. F.WETTENGEL, J.HOFFMANN, P. C.STAFFORST, T.: "Optimal guideRNAs for re-directing deaminase activity of hADAR1 and hADAR2 in trans", NUCLEIC ACIDS RES, vol. 42, 2014, pages e87, XP055260764, DOI: 10.1093/nar/gku272
SIMHADRI, V. L. ET AL.: "Prevalence of Pre-existing Antibodies to CRISPR-Associated Nuclease Cas9 in the USA Population", MOL THER METHODS CLIN DEV, vol. 10, 2018, pages 105 - 112, XP055569070, DOI: 10.1016/j.omtm.2018.06.006
SINNAMON, J. R. ET AL.: "Site-directed RNA repair of endogenous Mecp2 RNA in neurons", PROC NATL ACAD SCI U S A, vol. 114, 2017, pages E9395 - E9402, XP055588475, DOI: 10.1073/pnas.1715320114
TAN, M. H.: "Dynamic landscape and regulation of RNA editing in mammals", NATURE, vol. 550, 2017, pages 249 - 254
TEOH, P. J. ET AL.: "Aberrant hyperediting of the myeloma transcriptome by ADAR1 confers oncogenicity and is a marker of poor prognosis", BLOOD, vol. 132, 2018, pages 1304 - 1317, XP086685879, DOI: 10.1182/blood-2018-02-832576
TIJSSEN: "Laboratory Techniques In Biochemistry And Molecular Biology- Hybridization With Nucleic Acid Probes Part I", 1993, ELSEVIER, article "Overview of principles of hybridization and the strategy of nucleic acid probe assay"
VALLECILLO-VIEJO, I. C.LISCOVITCH-BRAUER, N.MONTIEL-GONZALEZ, M. F.EISENBERG, E.ROSENTHAL, J. J. C.: "Abundant off-target edits from site-directed RNA editing can be reduced by nuclear localization of the editing enzyme", RNA BIOLOGY, vol. 15, 2018, pages 104 - 114, XP055614862, DOI: 10.1080/15476286.2017.1387711
VAN DER AUWERA, G. A. ET AL.: "From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline", CURR PROTOC BIOINFORMATICS, vol. 43, no. 11, 2013, pages 11 - 33
VOGEL, P. ET AL.: "Efficient and precise editing of endogenous transcripts with SNAP-tagged ADARs", NATMETHODS, vol. 15, 2018, pages 535 - 538, XP036542160, DOI: 10.1038/s41592-018-0017-z
VOGEL, P., SCHNEIDER, M. F., WETTENGEL, J., STAFFORST, T.: " Improving site-directed RNA editing in vitro and in cell culture by chemical modification of the guideRNA", ANGEWANDTE CHEMIE, vol. 53, 2014, pages 6267 - 6271, XP055763014, DOI: 10.1002/anie.201402634
VOGEL, P.HANSWILLEMENKE, A.STAFFORST, T.: "Switching Protein Localization by Site-Directed RNA Editing under Control of Light", ACS SYNTHETIC BIOLOGY, vol. 6, 2017, pages 1642 - 1649
WAGNER, D. L. ET AL.: "High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population", NATMED, vol. 25, 2019, pages 242 - 248
WAGNER, R. W. ET AL.: "Double-stranded RNA unwinding and modifying activity is detected ubiquitously in primary tissues and cell lines", MOL CELL BIOL, vol. 10, 1990, pages 5586 - 5590
WAHLSTEDTHELENEMARIE: "Site-selective versus promiscuous A-to-I editing", WILEY INTERDISCIPLINARY REVIEWS: RNA, vol. 2, no. 6, 2011, pages 761 - 771
WANG, K.LI, M.HAKONARSON, H.: "ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data", NUCLEIC ACIDS RES, vol. 38, 2010, pages e164, XP055596024, DOI: 10.1093/nar/gkq603
WETTENGEL, J., REAUTSCHNIG, P., GEISLER, S., KAHLE, P. J., STAFFORST, T.: "Harnessing human ADAR2 for RNA repair - Recoding a PINK1 mutation rescues mitophagy", NUCLEIC ACIDS RES, vol. 45, 2017, pages 2797 - 2808
WOOLF, T. M.CHASE, J. M.STINCHCOMB, D. T.: "Toward the therapeutic editing of mutated RNA sequences", PROC NATL ACAD SCI U S A, vol. 92, 1995, pages 8298 - 8302, XP000574995, DOI: 10.1073/pnas.92.18.8298
WOOLF, T.M . ET AL.: "Toward the therapeutic editing of mutated RNA sequences", PROC. NATL. ACAD. SCI. USA, vol. 92, 31 August 1995 (1995-08-31), pages 8298 - 8302, XP000574995, DOI: 10.1073/pnas.92.18.8298 *
YIN, H., KAUFFMAN, K. J., ANDERSON, D. G.: "Delivery technologies for genome editing", NAT REV DRUG DISCOV, vol. 16, 2017, pages 387 - 399, XP055426482, DOI: 10.1038/nrd.2016.280
ZHENG, Y.LORENZO, C.BEAL, P. A.: "DNA editing in DNA/RNA hybrids by adenosine deaminases that act on RNA", NUCLEIC ACIDS RES, vol. 45, 2017, pages 3369 - 3377, XP055404026, DOI: 10.1093/nar/gkx050
ZHOU, Y.ZHANG, H.WEI, W.: "Simultaneous generation of multi-gene knockouts in human cells", FEBS LETT, vol. 590, 2016, pages 4343 - 4353, XP055570531, DOI: 10.1002/1873-3468.12469
ZUO, E. ET AL.: "Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos", SCIENCE, 2019

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11608503B2 (en) 2017-03-03 2023-03-21 The Regents Of The University Of California RNA targeting of mutations via suppressor tRNAs and deaminases
US11932856B2 (en) 2017-03-03 2024-03-19 The Regents Of The University Of California RNA targeting of mutations via suppressor tRNAs and deaminases
US11479775B2 (en) 2017-03-03 2022-10-25 The Regents Of The University Of California RNA targeting of mutations via suppressor tRNAs and deaminases
US11702658B2 (en) 2019-04-15 2023-07-18 Edigene Therapeutics (Beijing) Inc. Methods and compositions for editing RNAs
US11661596B2 (en) 2019-07-12 2023-05-30 Peking University Targeted RNA editing by leveraging endogenous ADAR using engineered RNAs
US11827880B2 (en) 2019-12-02 2023-11-28 Shape Therapeutics Inc. Therapeutic editing
WO2021136408A1 (zh) 2019-12-30 2021-07-08 博雅辑因(北京)生物科技有限公司 一种基于leaper技术治疗mps ih的方法和组合物
WO2021136404A1 (zh) 2019-12-30 2021-07-08 博雅辑因(北京)生物科技有限公司 一种治疗Usher综合征的方法和其组合物
WO2021209010A1 (zh) * 2020-04-15 2021-10-21 博雅辑因(北京)生物科技有限公司 一种治疗赫勒氏综合征的方法和药物
WO2021243023A1 (en) * 2020-05-28 2021-12-02 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of serpina1
CN113897359A (zh) * 2020-07-06 2022-01-07 博雅辑因(北京)生物科技有限公司 一种改善的rna编辑方法
WO2022007803A1 (zh) 2020-07-06 2022-01-13 博雅辑因(北京)生物科技有限公司 一种改善的rna编辑方法
WO2022091100A1 (en) 2020-10-29 2022-05-05 Yeda Research And Development Co. Ltd. Polynucleotides for rna editing and methods of using same
WO2022147573A1 (en) * 2021-01-04 2022-07-07 The Regents Of The University Of California Programmable rna editing in vivo via recruitment of endogenous adars
WO2023278436A1 (en) * 2021-06-29 2023-01-05 Shape Therapeutics Inc. Engineered guide rnas and polynucleotides
WO2023020574A1 (en) * 2021-08-18 2023-02-23 Peking University Engineered adar-recruiting rnas and methods of use thereof
WO2023077135A1 (en) * 2021-10-29 2023-05-04 Duke University Compositions and systems for rna-programmable cell editing and methods of making and using same
WO2024086650A1 (en) * 2022-10-18 2024-04-25 Locanabio, Inc. Compositions and methods comprising programmable snrnas for rna editing

Also Published As

Publication number Publication date
EP4306116A2 (en) 2024-01-17
BR112021006844A8 (pt) 2023-03-21
CA3115864A1 (en) 2020-04-16
TW202028466A (zh) 2020-08-01
EP3864152A4 (en) 2021-10-13
EP4306116A3 (en) 2024-04-17
EP3864152A1 (en) 2021-08-18
SG11202103666PA (en) 2021-05-28
PH12021550805A1 (en) 2021-10-04
CO2021005956A2 (es) 2021-07-30
CN115651927A (zh) 2023-01-31
AU2023202184A1 (en) 2023-05-04
KR102666695B1 (ko) 2024-05-23
JP2023078373A (ja) 2023-06-06
MX2021004187A (es) 2021-09-08
IL282191A (en) 2021-05-31
BR112021006844A2 (pt) 2021-07-20
JP7252328B2 (ja) 2023-04-04
CL2021000880A1 (es) 2021-12-10
US20210310026A1 (en) 2021-10-07
CN115651927B (zh) 2023-06-16
US20210355494A1 (en) 2021-11-18
PE20211402A1 (es) 2021-08-02
CN116218836A (zh) 2023-06-06
DK3864152T3 (da) 2023-10-09
AU2019357450B2 (en) 2023-01-12
ECSP21033637A (es) 2021-08-31
AU2019357450A1 (en) 2021-06-03
CN113631708B (zh) 2022-05-24
CN113631708A (zh) 2021-11-09
CN116042611A (zh) 2023-05-02
EP3864152B1 (en) 2023-08-16
CR20210243A (es) 2021-10-07
KR20210076082A (ko) 2021-06-23
JP2021534810A (ja) 2021-12-16
ES2962434T3 (es) 2024-03-19

Similar Documents

Publication Publication Date Title
AU2019357450B2 (en) Methods and compositions for editing RNAs
US11702658B2 (en) Methods and compositions for editing RNAs
US11661596B2 (en) Targeted RNA editing by leveraging endogenous ADAR using engineered RNAs
EP3589751A1 (en) RNA TARGETING OF MUTATIONS VIA SUPPRESSOR tRNAs AND DEAMINASES
WO2022150974A1 (en) Targeted rna editing by leveraging endogenous adar using engineered rnas
Qu et al. Leveraging endogenous ADAR for programmable editing on RNA

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19870299

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3115864

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021520227

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021006844

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217014349

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019870299

Country of ref document: EP

Effective date: 20210512

ENP Entry into the national phase

Ref document number: 2019357450

Country of ref document: AU

Date of ref document: 20191012

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021006844

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210409